EP1781303A1 - Procedes et reactifs pour le traitement de troubles metaboliques - Google Patents

Procedes et reactifs pour le traitement de troubles metaboliques

Info

Publication number
EP1781303A1
EP1781303A1 EP05768186A EP05768186A EP1781303A1 EP 1781303 A1 EP1781303 A1 EP 1781303A1 EP 05768186 A EP05768186 A EP 05768186A EP 05768186 A EP05768186 A EP 05768186A EP 1781303 A1 EP1781303 A1 EP 1781303A1
Authority
EP
European Patent Office
Prior art keywords
inhibitors
composition
insulin
analog
antagonists
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP05768186A
Other languages
German (de)
English (en)
Other versions
EP1781303A4 (fr
Inventor
Margaret S. Lee
Grant R. Zimmerman
Alyce Lynn Finelli
Daniel Grau
Curtis Keith
M. James Nichols
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zalicus Inc
Original Assignee
CombinatoRx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=35783202&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP1781303(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by CombinatoRx Inc filed Critical CombinatoRx Inc
Publication of EP1781303A1 publication Critical patent/EP1781303A1/fr
Publication of EP1781303A4 publication Critical patent/EP1781303A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/603Salicylic acid; Derivatives thereof having further aromatic rings, e.g. diflunisal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/28Mercury; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the invention relates to the treatment, prevention, and reduction of metabolic disorders, such as diabetes and obesity.
  • metabolic disorders such as diabetes and obesity.
  • insulin is secreted and stimulates cells of the peripheral tissues (skeletal muscles and fat) to actively take up glucose from the blood as a source of energy.
  • Loss of glucose homeostasis as a result of faulty insulin secretion or action typically results in metabolic disorders such as diabetes, which may be co-triggered or further exacerbated by obesity. Because these conditions are often fatal, strategies to restore adequate glucose clearance from the bloodstream are required.
  • diabetes may arise secondary to any condition that causes extensive damage to the pancreas (e.g., pancreatitis, tumors, administration of certain drugs such as corticosteroids or pentamidine, iron overload (e.g., hemochromatosis), acquired or genetic endocrinopathies, and surgical excision), the most common forms of diabetes typically arise from primary disorders of the insulin signaling system.
  • type 1 diabetes also known as insulin dependent diabetes (IDDM)
  • type 2 diabetes also known as insulin independent or non-insulin dependent diabetes (NIDDM)
  • Type 1 diabetes which accounts for approximately 10% of all cases of primary diabetes, is an organ-specific autoimmune disease characterized by the extensive destruction of the insulin-producing beta cells of the pancreas. The consequent reduction in insulin production inevitably leads to the deregulation of glucose metabolism. While the administration of insulin provides significant benefits to patients suffering from this condition, the short serum half-life of insulin is a major impediment to the maintenance of normoglycemia. An alternative treatment is islet transplantation, but this strategy has been associated with limited success.
  • Type 2 diabetes which affects a larger proportion of the population, is characterized by a deregulation in the secretion of insulin and/or a decreased response of peripheral tissues to insulin, i.e., insulin resistance. While the pathogenesis of type 2 diabetes remains unclear, epidemiologic studies suggest that this form of diabetes results from a collection of multiple genetic defects or polymorphisms, each contributing its own predisposing risks and modified by environmental factors, including excess weight, diet, inactivity, drugs, and excess alcohol consumption. Although various therapeutic treatments are available for the management of type 2 diabetes, they are associated with various debilitating side effects. Accordingly, patients diagnosed with or at risk of having type 2 diabetes are often advised to adopt a healthier lifestyle, including loss of weight, change in diet, exercise, and moderate alcohol intake. Such lifestyle changes, however, are not sufficient to reverse the vascular and organ damages caused by diabetes.
  • the present invention features compositions, methods, and kits for treating, preventing, and reducing metabolic disorders.
  • This invention is particularly useful for treating patients having or at risk of having any condition that is characterized by a state of hyperglycemia, which may be caused, for example, by an alteration in the insulin signaling pathway (e.g., a reduction in insulin production, resistance to insulin, or both).
  • Exemplary disorders amenable to treatment according to this invention are obesity, diabetes (e.g., type 1 diabetes, type 2 diabetes, maturity- onset diabetes of the young (MODY), and gestational diabetes), satiety, endocrine deficiencies of aging, and any of their associated complications (e.g., Syndrome X, diabetic retinopathy, diabetic nephropathy, diabetic neuropathy, peripheral vascular disease, hyperlipidemia, hypertension, atherosclerosis, and coronary heart disease).
  • diabetes e.g., type 1 diabetes, type 2 diabetes, maturity- onset diabetes of the young (MODY), and gestational diabetes
  • satiety e.g., endocrine deficiencies of aging, and any of their associated complications (e.g., Syndrome X, diabetic retinopathy, diabetic nephropathy, diabetic neuropathy, peripheral vascular disease, hyperlipidemia, hypertension, atherosclerosis, and coronary heart disease).
  • the invention features a composition that includes (a) bezafibrate or an analog thereof; and (b) diflunisal or an analog thereof, wherein the bezafibrate and diflunisal are present in amounts that, when administered to a patient, are sufficient to treat, prevent, or reduce a metabolic disorder (e.g., diabetes or obesity).
  • a metabolic disorder e.g., diabetes or obesity
  • Exemplary combinations are bezafibrate and diflunisal; bezafibrate and bismuth subsalicylate; bezafibrate and nimosulide; bezafibrate and oxaprozin; bezafibrate and diclofenac; bezafibrate and sundilac; bezafibrate and ibuprofen; clofibrate and diflunisal; clofibrate and bismuth subsalicylate; clofibrate and nimosulide; clofibrate and oxaprozin; clofibrate and diclofenac; clofibrate and sundilac; clofibrate and ibuprofen; clofibrinc acid and diflunisal; clofibric acid and bismuth subsalicylate; clofibric acid and nimosulide; clofibric acid and bis
  • the invention features a composition that includes (a) bezafibrate or an analog thereof; and (b) cinnamic acid or an analog thereof, wherein the bezafibrate and difhmisal are present in amounts that, when administered to a patient, are sufficient to treat, prevent, or reduce a metabolic disorder.
  • Exemplary bezafibrate analogs are binifibrate, ciprofibrate, clinofibrate, clofibrate, clofibric acid, etofibrate, fenofibrate, or gemfibrozil.
  • the composition may include a third agent selected from the group consisting of sulfonylureas, non- sulfonylurea secretagogues, insulin, insulin analogs, glucagon-like peptides, exendin-4 polypeptides, beta 3 adrenoceptor agonists, PPAR agonists, dipeptidyl peptidase IV inhibitors, biguanides, alpha-glucosidase inhibitors, immunomodulators, statins and statin-containing combinations, angiotensin converting enzyme inhibitors, adenosine Al receptor agonists, adenosine A2 receptor agonists, aldosterone antagonists, alpha 1 adrenoceptor antagonists, alpha 2 adrenoceptor agonists, alpha 2 adrenoceptor agonists, angiotensin receptor antagonists, antioxidants, ATPase inhibitors, atrial peptide agonists,
  • the invention also features a method for treating, preventing, or reducing a metabolic disorder in a patient in need thereof by administering to the patient (i) bezafibrate or an analog thereof; and (ii) diflunisal or an analog thereof, wherein the bezafibrate and diflunisal are administered in amounts that together are sufficient to treat, prevent, or reduce a metabolic disorder.
  • Exemplary combinations are bezafibrate and diflunisal; bezafibrate and bismuth subsalicylate; bezafibrate and nimosulide; bezafibrate and oxaprozin; bezafibrate and diclofenac; bezafibrate and sundilac; bezafibrate and ibuprofen; clofibrate and diflunisal; clofibrate and bismuth subsalicylate; clofibrate and nimosulide; clofibrate and oxaprozin; clofibrate and diclofenac; clofibrate and sundilac; clofibrate and ibuprofen; clofibrinc acid and diflunisal; clofibric acid and bismuth subsalicylate; clofibric acid and nimosulide; clofibric acid and bis
  • the invention also features a method for treating, preventing, or reducing a metabolic disorder in a patient in need thereof by administering to the patient (i) bezafibrate or an analog thereof; and (ii) cinnamic acid or an analog thereof, wherein the bezafibrate and cinnamic acid are administered in amounts that together are sufficient to treat, prevent, or reduce a metabolic disorder.
  • the patient may also be administered a third agent selected from the group consisting of sulfonylureas, non-sulfonylurea secretagogues, insulin, insulin analogs, glucagon-like peptides, exendin-4 polypeptides, beta 3 adrenoceptor agonists, PPAR agonists, dipeptidyl peptidase IV inhibitors, biguanides, alpha-glucosidase inhibitors, immunomodulators, statins and statin-containing combinations, angiotensin converting enzyme inhibitors, adenosine Al receptor agonists, adenosine A2 receptor agonists, aldosterone antagonists, alpha 1 adrenoceptor antagonists, alpha 2 adrenoceptor agonists, alpha 2 adrenoceptor agonists, angiotensin receptor antagonists, antioxidants, ATPase inhibitors, atrial peptide agonists
  • a third agent selected from
  • the invention also features a kit that includes (i) bezafibrate or an analog thereof; and (ii) instructions for administering bezafibrate and cinnamic acid or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) bezafibrate or an analog thereof; and (ii) instructions for administering bezafibrate and diflunisal or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) diflunisal or an analog thereof; and (ii) instructions for administering diflunisal and bezafibrate or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) cinnamic acid or an analog thereof; and (ii) instructions for administering cinnamic acid and bezafibrate or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) a composition containing bezafibrate or an analog thereof and cinnamic acid or an analog thereof; and (ii) instructions for administering the composition to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) a composition containing bezafibrate or an analog thereof and diflunisal or an analog thereof; and (ii) instructions for administering the composition to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) bezafibrate or an analog thereof; (ii) cinnamic acid or an analog thereof; and (iii) instructions for administering bezafibrate and cinnamic acid to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) bezafibrate or an analog thereof; (ii) diflunisal or an analog thereof; and (iii) instructions for administering bezafibrate and diflunisal to a patient having or at risk of having a metabolic disorder.
  • the invention also features a composition that includes (a) a PPAR agonist; and (b) diflunisal or an analog thereof, wherein the PPAR agonist and diflunisal are present in amounts that, when administered to a patient, are sufficient to treat, prevent, or reduce a metabolic disorder.
  • the invention also features a composition that includes (a) a PPAR agonist; and (b) cinnamic acid or an analog thereof, wherein the PPAR agonist and diflunisal are present in amounts that, when administered to a patient, are sufficient to treat, prevent, or reduce a metabolic disorder.
  • the patient may also be administered a third agent selected from the group consisting of sulfonylureas, non-sulfonylurea secretagogues, insulin, insulin analogs, glucagon-like peptides, exendin-4 polypeptides, beta 3 adrenoceptor agonists, PPAR agonists, dipeptidyl peptidase IV inhibitors, biguanides, alpha-glucosidase inhibitors, immunomodulators, statins and statin-containing combinations, angiotensin converting enzyme inhibitors, adenosine Al receptor agonists, adenosine A2 receptor agonists, aldosterone antagonists, alpha 1 adrenoceptor antagonists, alpha 2 adrenoceptor agonists, alpha 2 adrenoceptor agonists, angiotensin receptor antagonists, antioxidants, ATPase inhibitors, atrial peptide agonists
  • a third agent selected from
  • the invention also features a method for treating, preventing, or reducing a metabolic disorder in a patient in need thereof by administering to the patient (i) a PPAR agonist; and (ii) diflunisal or an analog thereof, wherein the PPAR ⁇ agonist and diflunisal are administered in amounts that together are sufficient to treat, prevent, or reduce a metabolic disorder.
  • the invention also features a method for treating, preventing, or reducing a metabolic disorder in a patient in need thereof by administering to the patient (i) a PPAR agonist; and (ii) cinnamic acid or an analog thereof, wherein the PPAR ⁇ agonist and cinnamic acid are administered in amounts that together are sufficient to treat, prevent, or reduce a metabolic disorder.
  • the invention also features a kit that includes (i) a PPAR agonist; and (ii) instructions for administering the PPAR agonist and cinnamic acid or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention features a kit that includes (i) a PPAR agonist; and (ii) instructions for administering the PPAR agonist and diflunisal or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention features a kit that includes (i) diflunisal or an analog thereof; and (ii) instructions for administering diflunisal and a PPAR agonist or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) cinnamic acid or an analog thereof; and (ii) instructions for administering cinnamic acid and a PPAR agonist or an analog thereof to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) a composition containing a PPAR agonist or an analog thereof and cinnamic acid or an analog thereof; and (ii) instructions for administering the composition to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) a composition containing a PPAR agonist or an analog thereof and diflunisal or an analog thereof; and (ii) instructions for administering the composition to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) a PPAR agonist or an analog thereof; (ii) cinnamic acid or an analog thereof; and (iii) instructions for administering the PPAR agonist and cinnamic acid to a patient having or at risk of having a metabolic disorder.
  • the invention also features a kit that includes (i) a PPAR agonist or an analog thereof; (ii) diflunisal or an analog thereof; and (iii) instructions for administering the PPAR agonist and diflunisal to a patient having or at risk of having a metabolic disorder.
  • the PPAR agonist is desirably a PPAR ⁇ agonist (e.g., balaglitazone, troglitazone, pioglitazone, ciglitazone, englitazone, rosiglitazone, darglitazone, englitazone, netoglitazone, KRP-297, JTT-501, NC- 2100, NIP-223, MCC-555, L-764486, CS-011, GI262570, GW347845, or FK614).
  • the invention also features a method of treating a metabolic disorder by administering to a mammal (e.g., human) one or more agents listed in Table 1 in an amount sufficient to treat, prevent, or reduce the metabolic disorder.
  • the mammal being treated may be administered two agents listed in Table 1 within 28 days of each other in amounts that together are sufficient to treat, prevent, or reduce the metabolic disorder.
  • the two agents are desirably administered within 14 days of each other, more desirably within seven days of each other, and even more desirably within twenty-four hours of each other, or even simultaneously (i.e., concomitantly). If desired, either one of the two agents may be administered in low dosage.
  • the mammal being treated may receive an additional therapeutic regimen.
  • a therapeutic agent is employed as the additional therapeutic regimen, the agent or agents from Table 1 and the additional agent are present in amounts that, when administered to a mammal, are together sufficient to treat, prevent, or reduce a metabolic disorder.
  • the additional agent may be selected from the group consisting of sulfonylureas, non-sulfonylurea secretagogues, insulin, insulin analogs, glucagon-like peptides, exendin-4 polypeptides, beta 3 adrenoceptor agonists, PPAR agonists, dipeptidyl peptidase IV inhibitors, biguanides, alpha- glucosidase inhibitors, immunomodulators, statins and statin-containing combinations, angiotensin converting enzyme inhibitors, adenosine Al receptor agonists, adenosine A2 receptor agonists, aldosterone antagonists, alpha 1 adrenoceptor antagonists, alpha 2 adrenoceptor agonists, alpha 2 adrenoceptor agonists, angiotensin receptor antagonists, antioxidants, ATPase inhibitors, atrial peptide agonists, beta adrenoceptor antagonists,
  • more than one therapeutic agent may be used with any of the agents listed in Table 1.
  • the different agents may be admixed together in a single formulation.
  • the agents may be administered simultaneously or within 14 days, 7 days, or 1 day of each other.
  • These agents may or may not be administered by the same route of administration (e.g., oral, intravenous, intramuscular, ophthalmic, topical, dermal, sub-cutaneous, and rectal).
  • the additional therapeutic regimen may involve a lifestyle change, including the adoption of a low-fat diet or low-sodium diet, stress management, physical exercise, reduction in alcohol intake, or reduction in smoking.
  • the present invention features a kit that includes any one of the agents listed in Table 1 and instructions for its administration to a patient having or at risk of having a metabolic disorder.
  • the kit contains two, three, four, or more than four agents from Table 1 that may or may not be admixed in the same formulation.
  • This kit may also contain instructions for administering this agent with a second agent listed in Table 1.
  • the invention also features a kit that includes (a) one, two, three, or more agents listed in Table 1 and (b) one or more of the following agents: a sulfonylurea, non- sulfonylurea secretagogue, insulin, insulin analog, glucagon-like peptide, exendin-4, YM 178, FK614, dipeptidyl peptidase IV inhibitor, biguanide, thiazalidinedione, alpha-glucosidase inhibitor, immunosuppressant, immmunomodulator, angiotensin converting enzyme (ACE) inhibitor, angiotensin II receptor blocker, or antioxidant.
  • a sulfonylurea non- sulfonylurea secretagogue
  • insulin insulin analog
  • glucagon-like peptide exendin-4
  • YM 178 exendin-4
  • FK614 dipeptidyl peptidase IV inhibitor
  • biguanide
  • the kit also includes instructions for administering these agents to a patient having or at risk of having a metabolic disorder.
  • the kit of the invention may contain one, two, three, or more agent listed in Table 1 or any one of the following agents: a sulfonylurea, non- sulfonylurea secretagogue, insulin, insulin analog, glucagon-like peptide, exendin- 4, YM178, FK614, dipeptidyl peptidase IV inhibitor, biguanide, thiazalidinedione, alpha-glucosidase inhibitor, immunosuppressant, immmunomodulator, angiotensin converting enzyme (ACE) inhibitor, angiotensin II receptor blocker, and antioxidant, as well as instructions for administering these two agents together to a patient having or at risk of having a metabolic disorder.
  • ACE angiotensin converting enzyme
  • the invention also features a method of identifying a combination of agents useful for the treatment, prevention, or reduction of a metabolic disorder, involving the steps of: (a) contacting cells with an agent listed in Table 1 and a candidate compound; and (b) determining whether this combination of agents reduces glucose levels relative to cells contacted only with the agent from Table 1 but not contacted with the candidate compound.
  • a reduction in glucose levels identifies the combination as being useful for the treatment, prevention, or reduction of a metabolic disorder.
  • Glucose levels may be reduced, for example, by altering insulin signaling (therefore increasing glucose uptake into cells and subsequent storage or metabolism, for example), altering glucose transporter activity (e.g., increasing GLUT4 expression, translocation, or intrinsic activity), increasing the amount of insulin-sensitive tissue (e.g., by increasing adipocyte or muscle cell differentiation), or altering gene transcription in adipocytes or muscle cells (e.g., altered secretion of factors from adipocytes and expression of metabolic pathway genes).
  • altering insulin signaling (therefore increasing glucose uptake into cells and subsequent storage or metabolism, for example), altering glucose transporter activity (e.g., increasing GLUT4 expression, translocation, or intrinsic activity), increasing the amount of insulin-sensitive tissue (e.g., by increasing adipocyte or muscle cell differentiation), or altering gene transcription in adipocytes or muscle cells (e.g., altered secretion of factors from adipocytes and expression of metabolic pathway genes).
  • the screening method for identifying a useful therapeutic combination involves the steps of: (a) contacting cells with an agent listed in Table 1 and a candidate compound; and (b) determining whether this combination of agents alters insulin signaling such that glucose levels are reduced relative to cells contacted with the agent from Table 1 but not contacted with the candidate compound.
  • An alteration in insulin signaling that reduces glucose levels identifies the combination as being useful for the treatment, prevention, or reduction of a metabolic disorder.
  • Mammalian (e.g., human) cells may be employed in any of the screening methods described herein. Particularly useful cells are muscle cells, intestinal cells, adipocytes, liver cells, and pancreatic cells. Optionally, these cells have an alteration in insulin signaling activity such that glucose levels are increased. Such a reduction in glucose levels may result from an increase in insulin production, an increase in insulin secretion, an increase in glucose uptake by peripheral tissues, a reduction in hepatic glucose production, or a reduction in the absorption of carbohydrates from the intestines.
  • Analogs of any of the compounds listed in Table 1 may be used in any of the methods, kits, and compositions of the invention. Such analogs include any agent from the same chemical class, mechanistic class, or therapeutic class as those listed in Table 2.
  • treating, reducing, or preventing a metabolic disorder is meant ameliorating such a condition before or after it has occurred.
  • reduction or degree of prevention is at least 5%, 10%, 20%, 40%, 50%, 60%, 80%, 90%, 95%, or 100% as measured by any standard technique.
  • a patient who is being treated for a metabolic disorder is one who a medical practitioner has diagnosed as having such a condition. Diagnosis may be performed by any suitable means, such as those described herein.
  • a patient in whom the development of diabetes or obesity is being prevented may or may not have received such a diagnosis.
  • patients of the invention may have been subjected to standard tests or may have been identified, without examination, as one at high risk due to the presence of one or more risk factors, such as family history, obesity, particular ethnicity (e.g., African Americans and Hispanic Americans), gestational diabetes or delivering a baby that weighs more than nine pounds, hypertension, having a pathological condition predisposing to obesity or diabetes, high blood levels of triglycerides, high blood levels of cholesterol, presence of molecular markers (e.g., presence of autoantibodies), and age (over 45 years of age).
  • An individual is considered obese when their weight is 20% (25% in women) or more over the maximum weight desirable for their height.
  • An adult who is more than 100 pounds overweight, is considered to be morbidly obese.
  • Obesity is also defined as a body mass index (BMI) over 30 kg/m 2 .
  • a metabolic disorder any pathological condition resulting from an alteration in a patient's metabolism. Such disorders include those resulting from an alteration in glucose homeostasis resulting, for example, in hyperglycemia.
  • an alteration in glucose levels is typically an increase in glucose levels by at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or even 100% relative to such levels in a healthy individual.
  • Metabolic disorders include obesity and diabetes (e.g., diabetes type I, diabetes type II, MODY, and gestational diabetes), satiety, and endocrine deficiencies of aging.
  • reducing glucose levels is meant reducing the level of glucose by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% relative to an untreated control.
  • glucose levels are reduced to normoglycemic levels, i.e., between 150 to 60 mg/dL, between 140 to 70 mg/dL, between 130 to 70 mg/dL, between 125 to 80 mg/dL, and preferably between 120 to 80 mg/dL.
  • Such reduction in glucose levels may be obtained by increasing any one of the biological activities associated with the clearance of glucose from the blood. Accordingly, an agent having the ability to reduce glucose levels may increase insulin production, secretion, or action.
  • Insulin action may be increased, for example, by increasing glucose uptake by peripheral tissues and/or by reducing hepatic glucose production.
  • the agent of the invention may reduce the absorption of carbohydrates from the intestines, alter glucose transporter activity (e.g., by increasing GLUT4 expression, intrinsic activity, or translocation), increase the amount of insulin-sensitive tissue (e.g., by increasing muscle cell or adipocyte cell differentiation), or alter gene transcription in adipocytes or muscle cells (e.g., altered secretion of factors from adipocytes expression of metabolic pathway genes).
  • the agent of the invention increases more than one of the activities associated with the clearance of glucose.
  • alter insulin signaling pathway such that glucose levels are reduced is meant to alter (by increasing or reducing) any one of the activities involved in insulin signaling such that the overall result is an increase in the clearance of glucose from plasma.
  • the agent of the invention alters the insulin signaling pathway causing an increase in insulin production, secretion, or action, an increase in glucose uptake by peripheral tissues, a reduction in hepatic glucose production, or a reduction in the absorption of carbohydrates from the intestines.
  • patient is meant any animal (e.g., a human), including horses, dogs, cats, pigs, goats, rabbits, hamsters, monkeys, guinea pigs, rats, mice, lizards, snakes, sheep, cattle, fish, and birds.
  • an amount sufficient is meant the amount of a compound, alone or in combination with another therapeutic regimen, required to treat, prevent, or reduce a metabolic disorder such as diabetes in a clinically relevant manner.
  • a sufficient amount of an active compound used to practice the present invention for therapeutic treatment of conditions caused by or contributing to diabetes varies depending upon the manner of administration, the age, body weight, and general health of the mammal or patient. Ultimately, the prescribers will decide the appropriate amount and dosage regimen. Additionally, an effective amount may be an amount of compound in the combination of the invention that is safe and efficacious in the treatment of a patient having a metabolic disorder such as diabetes over each agent alone as determined and approved by a regulatory authority (such as the U.S. Food and Drug Administration).
  • a treatment exhibits greater efficacy, or is less toxic, safer, more convenient, or less expensive than another treatment with which it is being compared. Efficacy may be measured by a skilled practitioner using any standard method that is appropriate for a given indication.
  • a “low dosage” is meant at least 5% less (e.g., at least 10%, 20%, 50%,
  • a low dosage of an agent that reduces glucose levels and that is formulated for administration by inhalation will differ from a low dosage of the same agent formulated for oral administration.
  • a “high dosage” is meant at least 5% (e.g., at least 10%, 20%, 50%, 100%, 200%, or even 300%) more than the highest standard recommended dosage of a particular compound for treatment of any human disease or condition.
  • corticosteroid any naturally occurring or synthetic compound characterized by a hydrogenated cyclopentanoperhydrophenanthrene ring system and having immunosuppressive and/or anti-inflammatory activity.
  • Naturally occurring corticosteroids are generally produced by the adrenal cortex. Synthetic corticosteroids may be halogenated. Exemplary corticosteroids are provided herein.
  • non-steroidal immunophilin-dependent immunosuppressant or “NsIDI” is meant any non-steroidal agent that decreases proinflammatory cytokine production or secretion, binds an immunophilin, or causes a down regulation of the proinflammatory reaction.
  • NsIDIs include calcineurin inhibitors, such as cyclosporine, tacrolimus, ascomycin, pimecrolimus, as well as other agents (peptides, peptide fragments, chemically modified peptides, or peptide mimetics) that inhibit the phosphatase activity of calcineurin.
  • NsIDIs also include rapamycin (sirolimus) and everolimus, which bind to an FK506-binding protein, FKBP- 12, and block antigen-induced proliferation of white blood cells and cytokine secretion.
  • small molecule immunomodulator is meant a non-steroidal, non- NsIDI compound that decreases proinflammatory cytokine production or secretion, causes a down regulation of the proinflammatory reaction, or otherwise modulates the immune system in an immunophilin-independent manner.
  • the number of atoms of a particular type in a substituent group is generally given as a range, e.g., an alkyl group containing from 1 to 4 carbon atoms or Cj_ 4 alkyl. Reference to such a range is intended to include specific references to groups having each of the integer number of atoms within the specified range.
  • an alkyl group from 1 to 4 carbon atoms includes each of C 1 , C 2 , C 3 , and C 4 .
  • a Q_ 12 heteroalkyl for example, includes from 1 to 12 carbon atoms in addition to one or more heteroatoms. Other numbers of atoms and other types of atoms may be indicated in a similar manner.
  • alkyl and the prefix “alk-” are inclusive of both straight chain and branched chain groups and of cyclic groups, i.e., cycloalkyl.
  • Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 6 ring carbon atoms, inclusive.
  • Exemplary cyclic groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups.
  • C 1--4 alkyl is meant a branched or unbranched hydrocarbon group having from 1 to 4 carbon atoms.
  • a C 1 ⁇ . alkyl group may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • C ⁇ 4 alkyls include, without limitation, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, and cyclobutyl.
  • halogen is meant bromine, chlorine, iodine, or fluorine.
  • alkoxy is meant a chemical substituent of the formula -OR, wherein R is selected from Ci_ 7 alkyl, C 2 _ 7 alkenyl, C 2 _ 7 alkynyl, C 2 _ 6 heterocyclyl, C 6 _ 12 aryl, C 7 _i 4 alkaryl, C 3 - ⁇ alkheterocyclyl, or Ci_ 7 heteroalkyl.
  • FIG 1 is a graph showing insulin sensitivity in male Sprague Dawley rats, as determined using the homeostasis model assessment (HOMA).
  • Insulin resistance was induced by four weeks of high fat feeding (60% of calories derived from fat). Drug treatment began one week after initiation of high fat diet. Drugs were administered daily, by oral gavage for a three week period. Following the three weeks of treatment, animals were fasted for five hours and anesthetized, and blood collected from the inferior vena cava for determination of serum glucose and insulin levels. Insulin sensitivity was determined using HOMA.
  • HOMA fasting serum glucose X fasting serum insulin
  • FIG 2 is a graph showing blood glucose levels in male Sprague Dawley rats. Insulin resistance was induced by four weeks of high fat feeding (60% of calories derived from fat). Drug treatment began one week after initiation of high fat diet. Drugs were administered daily, by oral gavage for a three week period. Following the three weeks of treatment, animals were fasted for five hours and anesthetized, and blood collected from the inferior vena cava.
  • FIG 3 is a graph showing blood serum insulin levels in male Sprague Dawley rats. Insulin resistance was induced by four weeks of high fat feeding
  • Drug treatment began one week after initiation of high fat diet. Drugs were administered daily, by oral gavage for a three week period. Following the three weeks of treatment, animals were fasted for five hours and anesthetized, and blood collected from the inferior vena cava.
  • the ability of the agent to cause the clearance of glucose may be attributed, for example, to its ability to increase insulin production, secretion, or action (e.g., stimulation of glucose uptake by peripheral tissues and/or reduction in hepatic glucose production), reduce the absorption of carbohydrates from the intestines, alter glucose transporter activity (e.g., by increasing GLUT4 expression, intrinsic activity, or translocation), increase the level of insulin-sensitive tissue (e.g., by increasing adipocyte or muscle cell differentiation), or alter gene transcription in adipocytes or muscle cells (e.g., altered secretion of factors from adipocytes and expression of metabolic pathway genes).
  • alter glucose transporter activity e.g., by increasing GLUT4 expression, intrinsic activity, or translocation
  • increase the level of insulin-sensitive tissue e.g., by increasing adipocyte or muscle cell differentiation
  • gene transcription in adipocytes or muscle cells e.g., altered secretion of factors from adipocytes and expression of metabolic pathway genes.
  • the administration of bezafibrate and cinnamic acid to a patient having a metabolic disorder such as diabetes within 14 days of each other will treat, prevent, or reduce the metabolic disorder.
  • the administration of bezafibrate and diflunisal to the patient within 14 days of each other will also treat, prevent, or reduce the metabolic disorder.
  • the two agents are desirably administered within 10 days of each other, more desirably within seven days of each other, and even more desirably within twenty- four hours of each other, one hour of each other, or even simultaneously (i.e., concomitantly). If desired, either one of the two agents may be administered in low dosage.
  • the aforementioned combinations of drugs can be used in a variety of compositions, methods, and kits, as described herein. Additionally, if desired, structural or functional analogs may be used in place of one or more of the drugs in a combination. Such analogs are described below.
  • compounds that, alone or in combination, are effective in the treatment, reduction, or prevention of metabolic disorders such as diabetes and obesity. Accordingly, a mammal that has been diagnosed with or is at risk of having a metabolic disorder is administered one, two, three, or more agents from Table 1. Optionally, analogs of these agents may be employed.
  • such administration may reduce the levels of glucose, reduce levels of LDL-cholesterol, increase the levels of HDL- cholesterol, result in a more favorable ratio between LDL-cholesterol and HDL- cholesterol, reduce the triglyceride values, or reduce serum levels of CRP (C- reactive protein).
  • CRP C- reactive protein
  • the ability of the agent to cause the clearance of glucose may be attributed, for example, to its ability to increase insulin production, secretion, or action (e.g., stimulation of glucose uptake by peripheral tissues and/or reduction in hepatic glucose production), reduce the absorption of carbohydrates from the intestines, alter glucose transporter activity (e.g., by increasing GLUT4 expression, intrinsic activity, or translocation), increase the level of insulin- sensitive tissue (e.g., by increasing adipocyte or muscle cell differentiation), or alter gene transcription in adipocytes or muscle cells (e.g., altered secretion of factors from adipocytes and expression of metabolic pathway genes).
  • the mammal may also receive other therapeutic regimens.
  • the mammal being treated is administered two agents listed in Table 1 within 28 days of each other in amounts that together are sufficient to treat, prevent, or reduce the metabolic disorder.
  • the two agents are desirably administered within 14 days of each other, more desirably within seven days of each other, and even more desirably within twenty- four hours of each other, or even simultaneously (i.e., concomitantly). If desired, either one of the two agents may be administered in low dosage.
  • Diagnosis of metabolic disorders The methods and compositions of the present invention are useful for treating any patient that has been diagnosed with or is at risk of having a metabolic disorder, such as diabetes.
  • a patient in whom the development of a metabolic disorder (e.g., diabetes or obesity) is being prevented may or may not have received such a diagnosis.
  • patients of the invention may have been subjected to standard tests or may have been identified, without examination, as one at high risk due to the presence of one or more risk factors.
  • Diagnosis of metabolic disorders may be performed using any standard method known in the art, such as those described herein. Methods for diagnosing diabetes are described, for example, in U.S. Patent No. 6,537,806, hereby incorporated by reference.
  • Diabetes may be diagnosed and monitored using, for example, urine tests (urinalysis) that measure glucose and ketone levels (products of the breakdown of fat); tests that measure the levels of glucose in blood; glucose tolerance tests; and assays that detect molecular markers characteristic of a metabolic disorder in a biological sample (e.g., blood, serum, or urine) collected from the mammal (e.g., measurements of Hemoglobin Ale (HbAIc) levels in the case of diabetes).
  • urine tests urinalysis
  • ketone levels products of the breakdown of fat
  • tests that measure the levels of glucose in blood
  • glucose tolerance tests e.g., glucose tolerance tests
  • assays that detect molecular markers characteristic of a metabolic disorder in a biological sample e.g., blood, serum, or urine
  • HbAIc Hemoglobin Ale
  • Patients may be diagnosed as being at risk or as having diabetes if a random plasma glucose test (taken at any time of the day) indicates a value of 200 mg/dL or more, if a fasting plasma glucose test indicates a value of 126 mg/dL or more (after 8 hours), or if an oral glucose tolerance test (OGTT) indicates a plasma glucose value of 200 mg/dL or more in a blood sample taken two hours after a person has consumed a drink containing 75 grams of glucose dissolved in water.
  • the OGTT measures plasma glucose at timed intervals over a 3 -hour period.
  • the level of plasma glucose in a diabetic patient that has been treated according to the invention ranges between 160 to 60 mg/dL, between 150 to 70 mg/dL, between 140 to 70 mg/dL, between 135 to 80 mg/dL, and preferably between 120 to 80 mg/dL.
  • HbAIc hemoglobin AIc
  • a person without diabetes typically has an HbAIc value that ranges between 4% and 6%.
  • HbAIc value of a patient being treated according to the present invention is reduced to less than 9%, less than 7%, less than 6%, and most preferably to around 5%.
  • the HbAIc levels of the patient being treated are preferably lowered by 10%, 20%, 30%, 40%, 50%, or more relative to such levels prior to treatment.
  • Gestational diabetes is typically diagnosed based on plasma glucose values measured during the OGTT. Since glucose levels are normally lower during pregnancy, the threshold values for the diagnosis of diabetes in pregnancy are lower than in the same person prior to pregnancy. If a woman has two plasma glucose readings that meet or exceed any of the following numbers, she has gestational diabetes: a fasting plasma glucose level of 95 mg/dL, a 1-hour level of 180 mg/dL, a 2-hour level of 155 mg/dL, or a 3-hour level of 140 mg/dL.
  • Ketone testing may also be employed to diagnose type 1 diabetes. Because ketones build up in the blood when there is not enough insulin, they eventually accumulate in the urine. High levels of blood ketones may result in a serious condition called ketoacidosis.
  • HbAIc hemoglobin AIc
  • Bezafibrate Bezafibrate (2-[4-2-[(4-chlorobenzoyl)amino]-ethyl]phenozy]-2- methylpropanoic acid) has the following structure:
  • Bezafibrate analogs include binifibrate, ciprofibrate, clinofibrate, clofibrate, etofibrate, fenofibrate, and gemfibrozil. Other bezafibrate analogs are described by formula (I).
  • each Of R 1 and R 2 is, independently, hydrogen, halogen, C ]-4 alkyl, and C 1-4 alkoxy
  • each of R 3 and R 4 is, independently, hydrogen and Ci -4 alkyl
  • n is 1, 2, and 3
  • Z is hydroxyl and Ci -4 alkyl.
  • Specific bezafibrate analogs of formula (I) are alpha-[4-(2-methoxy-5- chlorobenzoylaminoethyl)-phenoxy-]isobutyric acid, alpha- [4-(4- methylbenzoylaminomethyl)-phenoxy]-isobutyric acid, alpha-[4-(2- methylbenzoylaminoethyl)-phenoxy]-isobutyric acid, and alpha-[4-(4- chlorobenzoylaminoethyl)-phenoxy]-proprionic acid.
  • PPAR agonists are ligand-activated transcription factors belonging to the nuclear receptor superfamily. Three different PPARs have been identified to date (PP ARa, PPAR ⁇ and PPAR ⁇ ) each displaying distinct tissue distribution pattern. PPARs are activated by natural ligands such as fatty acids and eicosanoids (leukotrienes, prostaglandins) and by pharmacological agonists such as fibrates, binding to PP ARa and glitazones, binding to PPAR ⁇ . Upon ligand activation, PPARs regulate the transcription of several genes.
  • ligands such as fatty acids and eicosanoids (leukotrienes, prostaglandins)
  • pharmacological agonists such as fibrates, binding to PP ARa and glitazones, binding to PPAR ⁇ .
  • PPARs heterodimerize with another nuclear receptor, the retinoid X receptor, and modify the transcription of target genes after binding to specific peroxisome proliferator response elements (PPRE).
  • PPRE peroxisome proliferator response elements
  • glitazones e.g., balaglitazone, troglitazone, pioglitazone, ciglitazone, englitazone, rosiglitazone, darglitazone, englitazone, netoglitazone, KRP-297, JTT-501, NC-2100, NIP-223, MCC-555, L-764486, and CS-Ol 1
  • thiazolidinediones can be used in combination with cinnamic acid, diflunisal, or an analog thereof, for treating a patient having a metabolic disorder.
  • tyrosine-based PPAR ⁇ modulators e.g., GI262570; [(S)-2-(2 ⁇ benzoylphenylamino)-3-[4-[2-(5-methyl-2-phenyl-2-oxazol-4- yl)ethoxy] ⁇ henyl] ⁇ ropionic acid, and GW347845 (Cobb et al., J Med Chem.
  • PPAR agonists that may be used in lieu of bezafibrate in the compositions, methods, and kits of the invention are AA- 10090, AD-5075, AMG- 131, ARH-049020, AVE-0847, AVE-8134, AY-31637, BAY-549801, bexarotene, BM-131246, BM-501050, CLX-0921, CLX-0940, DRF-10945, DRF-4832, E- 3030, farglitazar, fenofibrate/metfo ⁇ nin, GW-0072, GW-1929, GW-2570, GW- 409544, GW-409890, GW-501516, GW-5393, GW-590735, GW-7282, GW- 9578, KHP-101, KT-6207, L-764406, LF-200337, LG-101506, LR-90, LY- 465608, LY-510929, LY-518674
  • statins may be used in lieu of bezafibrate in the compositions, methods, and kits of the invention.
  • Exemplary statins and statin-containing combinations are acitemate, amlodipine/atorvastatin, atorvastatin, atorvastatin/torcetrapib, BAY102987, BAY X 2678, BB476, bervastatin, BMY21950, BMY22089, cerivastatin, colestolone, CP83101, crilvastatin, dalvastatin, DMP565, ezetimibe/simvastatin, fluvastatin, glenvastatin, L659699, L669262, mevastatin, nicotinic acid/lovastatin, nicotinic acid/simvastatin, P882222, P882284, PD134965, PD135022, pitavastatin, rosuvastatin,
  • NSAIDs may also be used in lieu of bezafibrate in the combinations, methods, and kits of the invention.
  • Suitable NSAIDs include A183827, ABT963, aceclofenac, acemetacin, acetyl salicylic acid, AHRl 0037, alclofenac, alminoprofen, ampiroxicam, amtolmetin guacil, apazone, aspirin, atliprofen methyl ester, AU8001, benoxaprofen, benzydamine flufenamate, bermoprofen, bezpiperylon, BF388, BF389, BIRL790, BMS347070, bromfenac, bucloxic acid, butibufen, BW755C, C53, C73, C85, carprofen, CBSl 108, celecoxib, CHF2003, chlorobiphenyl, choline magnesium trisalicylate, CHXl 08, ci
  • R 1 and R 2 each represent a hydrogen atom or a C 1-4 alkyl group
  • R 3 and R 4 each represent a hydrogen atom or may be combined together to form an additional chemical bond
  • X represents a hydroxyl group, a halogen atom, a straight or branched chain saturated or unsaturated C 1-4 alkyl group, a straight or branched chain saturated or unsaturated C 1-4 alkoxy group, a C 1-4 acyloxy group, or a C 3-6 cycloalkyl group
  • n is zero or an integer from 1 to 3, with the proviso that when n is 2 or 3, each X may be the same or different and that when two X's are commonly the alkyl or alkoxy group, both X's may be combined together to form a ring.
  • cinnamic acid analogs of formula (II) are hydrocinnamic acid, 2-, 3- and 4-methylhydrocinnamic acid, 2-, 3- and 4-ethylhydrocinnamic acid, 2-, 3- and 4-propylhydrocinnamic acid, 2-, 3- and 4-hydroxyhydrocinnamic acid, 2-, 3- and 4-methoxyhydrocinnamic acid, 2-, 3- and 4-ethoxyhydrocinnamic acid, 2-, 3- and 4-chlorohydrocinnamic acid, 2-, 3- and 4-bromohydrocinnamic acid, 2-, 3- and 4-fluorohydrocinnamic acid, 2, A-, 2,5- and 3,4-dimethylhydrocinnamic acid, 2,4-diethylhydrocinnamic acid, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5- dihydroxyhydrocimmamic acid, 2,3-, 2,4-,
  • Diflunisal (2',4'-difluoro-4-hydroxy-[l,r-biphenyl]-3-carboxylic acid) is an NSAID having the structure: Methods of making diflunisal are described in U.S. Patent No. 3,714,226.
  • each X is, independently, a halogen atom
  • Ri is selected from the group consisting of hydroxy, phenoxy, di(Ci-C 4 )alkylamino, di(Ci-C 4 )alkylamino (C 1 - C 4 )alkoxy
  • R 2 is selected from the group consisting of hydrogen and (C 1 - C 4 )alkanoyl (such as acetyl, propionyl and butyryl); and
  • R 3 is selected from the group consisting of hydrogen and methyl.
  • Suitable NSAIDs include Al 83827, ABT963, aceclofenac, acemetacin, acetyl salicylic acid, AHRl 0037, alclofenac, alminoprofen, ampiroxicam, amtolmetin guacil, apazone, aspirin, atliprofen methyl ester, AU8001, benoxaprofen, benzydamine flufenamate, bermoprofen, bezpiperylon, BF388, BF389, BIRL790, BMS347070, bromfenac, bucloxic acid, butibufen, BW755C, C53, C73, C85, carprofen,
  • the present invention involves the administration of an effective amount of one, two, three, four, or more agents listed in Table 1 to a mammal at risk of having or having a metabolic disorder, thereby treating, preventing, and reducing such a disorder.
  • the agent of the invention increases the clearance of glucose from the plasma by any mechanism thereby reducing glucose levels to normoglycemic levels.
  • a combination of the invention may alter the insulin signaling pathway, reduce the absorption of carbohydrates from the intestines, alter glucose transporter activity (e.g., by increasing GLUT4 expression, intrinsic activity, or translocation), increase the level of insulin- sensitive tissue (e.g., by increasing adipocyte or muscle cell differentiation), or alter gene transcription in adipocytes or muscle cells (e.g., altered secretion of factors from adipocytes expression of metabolic pathway genes).
  • the plasma glucose levels of the patient being treated according to the invention is stabilized such that glucose levels range between 60 and 150 mg/dL, between 70 and 140 mg/dL, between 80 and 130 mg/dL, and preferably between 80 and 120 mg/dL prior to a meal and between 90 mg/dL to 160 mg/dL, between 90 mg/dL to 150 mg/dL, and preferably between 90 to 140 mg/dL two hours after eating.
  • a particularly desirable result of treatment is a reduction of any of the symptoms associated with the metabolic disorder.
  • Analogs of any of the compounds listed in Table 1 may be used in any of the methods, kits, and compositions of the invention. Analogs are known in the art (e.g., as described herein). Acemetacin analogs are described in German patent DE 2234651 and U.S.P.N. 3,910952. Acenocoumarol analogs are described in U.S.P.N. 2,648,682. Acetaminophen analogs are described in U.S. P. N. 2,998,450 and German patent DE 453577. Acetazolamide analogs are described in U.S.P.N. 2,554,816. Acetohexamide analogs are described in GB 912789.
  • Acetyldigitoxin analogs are described in U.S.P.N. 2,776,963. Alachlor analogs are described in NL 6602564. Albuterol (Salbutamol) analogs are described in ZA 67 05591 and U.S.P.N. 3,644,353. Alprenolol analogs are described in NL 6605692 and NL 6612958. Ametryn analogs are described in CH 337019 and U.S.P.N. 3,558,622. Atrazine analogs are described in CH 342784, CH 342785, HU 149189, and FR 1317812. Azathioprine analogs are described in U.S.P.N.
  • Candesartan (e.g., cilexetil salt) analogs are described in EP 459136 and U.S.P.N. 5,196,444, captopril analogs are described in DE 2703828, U.S.P.N. 4,046,889, and U.S.P.N. 4,105,776, and carbamazepine analogs are described in U.S.P.N. 2,948,718.
  • Carbinoxamine (e.g., maleate salt) analogs are described in U.S.P.N. 2,606,195, U.S.P.N. 2,800,485, and GB 905993.
  • Carisoprodol analogs are described in U.S.P.N.
  • cefamandole (e.g., nafate salt) analogs are described in German patents DE 2018600 and DE 2312997, as well as U.S.P.N. 3,641,021 and U.S.P.N. 3,840,531.
  • Cefpodoxime Proxetil analogs are described in EP 49118 and U.S.P.N. 4,486,425.
  • Ciclopirox analogs are described in ZA 69 06039 and U.S.P.N. 3,883,545.
  • Clenbuterol analogs are described in ZA 67 05692 and U.S.P.N. 3,536,712.
  • Diacerein analogs are described in German patent DE 2711493, Japanese Patent JP Kokai 83 225015, U.S.P.N. 4,244,968, and U.S.P.N. 4,346,103.
  • Diclofenac (e.g., sodium salt) analogs are described in U.S.P.N. 3,558,690 and NL 6604752.
  • Dicloxacillin (e.g., sodium salt) analogs are described in GB 978299 and U.S.P.N. 3,239,507.
  • Diilunisal analogs are described in ZA 67 01021, FR 1522570, and U.S.P.N. 3,714,226. Dimenhydrinate analogs are described in U.S.P.N.
  • Enalaprilat analogs are described in EP 12401 and U.S.P.N. 4,374,829.
  • Ethopropazine (e.g., hydrochloride salt) analogs are described in U.S.P.N. 2,607,773.
  • Etoposide analogs are described in U.S.P.N. 3,524,844.
  • Evans Blue analogs are described in German patents DE 35341, DE 38802, DE 3949, DE 57327, and DE 75469.
  • Exemestane analogs are described in U.S.P.N. 4,808,616 and German patent DE 3622841.
  • Flunixin (e.g., meglumine salt) analogs are described in BE 679271, BE 812772, as well as U.S.P.N.
  • Leflunomide analogs are described in German patent DE 2854439 and U.S.P.N. 4,284,786.
  • Levocabastine (e.g., hydrochloride salt) analogs are described in EP 34415 and U.S.P.N. 4,369,184.
  • Loxapine (e.g., hydrochloride salt) analogs are described in NL 6406089 and U.S.P.N. 3,546,226 and U.S.P.N. 3,412,193.
  • Mebhydroline (e.g., 1,5-Naphthalenedisulfonate salt) analogs are described in U.S.P.N. 2,786,059.
  • Meclofenoxate analogs are described in FR M398.
  • Mefenamic Acid analogs are described in BE 605302 and U.S.P.N. 3,138,636.
  • Meloxicam analogs are described in U.S.P.N. 4,233,299.
  • Melphalan analogs are described in U.S.P.N. 3,032,584 and U.S.P.N. 3,032,585.
  • Mercaptoethanol analogs are described in U.S.P.N. 2,402,665 and U.S.P.N. 3,394,192.
  • Metaproterenol (e.g., hemisulfate salt) analogs are described in U.S.P.N. 3,341,594 and BE 611502.
  • Methacholine analogs are described in U.S.P.N. 2,040,146.
  • Methdilazine analogs are described in U.S.P.N. 2,945,855.
  • Methylergonovine (e.g., maleate salt) analogs are described in U.S.P.N. 2,265,207.
  • Nateglinide analogs are described in EP 196222 and U.S.P.N. 4,816,484.
  • Nefopam analogs are described in NL 6606390 and U.S.P.N. 3,830,803.
  • Nimesulide analogs are described in BE 801812 and U.S.P.N. 3,840,597.
  • Norepinephrine (e.g., bitartrate salt) analogs are described in U.S.P.N. 2,774,789.
  • Olanzapine analogs are described in EP 454436 and U.S.P.N. 5,229,382.
  • Oleandomycin analogs are described in U.S.P.N. 2,757,123 and U.S.P.N. 2,842,481.
  • Orphenadrine (e.g., citrate salt) analogs are described in U.S.P.N. 2,567,351and U.S.P.N. 2,991,225.
  • Oxaprozin analogs are described in FR 2001036, GB 1206403, and U.S.P.N. 3,578,671.
  • Oxybutynin (e.g., chloride salt) analogs are described in GB 940540.
  • Oxymetazoline (e.g., hydrochloride salt) analogs are described in German patent DE 1117588.
  • Pergolide (e.g., mesylate salt) analogs are described in U.S.P.N. 4,166,182.
  • Phenacemide analogs are described in U.S.P.N. 2,887,513.
  • Phensuximide analogs are described in U.S.P.N. 2,643,258.
  • Phenylbutazone analogs are described in U.S.P.N. 2,562,830 and GB 812449.
  • Phenylephrine analogs are described in U.S.P.N. 1,932,347 and U.S.P.N. 1,954,389.
  • Phenytoin analogs are described in U.S.P.N. 2,409,754. Prazosin (e.g., hydrochloride salt) analogs are described in GB 1156973, U.S.P.N. 3,511,836, and NL 7206067. Promethazine (e.g., hydrochloride salt) analogs are described in U.S.P.N. 2,530,451 and U.S.P.N. 2,607,773. Prostaglandin (e.g., Prostaglandin E) analogs are described in GB 851827, U.S.P.N. 3,598,858, NL 6505799, DE 2126127, U.S.P.N.
  • Rolipram analogs are described in BE 826923 and U.S.P.N. 4,193,926.
  • Succinylcholine analogs are described in AT 171411.
  • Sulfaguanidine analogs are described in U.S.P.N. 2,218,490, U.S.P.N. 2,229,784, U.S.P.N. 2,233,569, and GB 551524.
  • Sulfamethizole analogs are described in U.S.P.N. 2,447,702.
  • Suprofen analogs are described in U.S.P.N. 4,035,376. Telmisartan analogs are described in EP 502314.
  • Terbutaline (e.g., sulfate salt) analogs are described in BE 704932 and U.S.P.N. 3,937,838.
  • Tetrahydrozoline (e.g., hydrochloride salt) analogs are described in U.S.P.N. 2,731,471 and U.S.P.N. 2,842,478.
  • Tinidazole analogs are described in U.S.P.N. 3,376,311. Tioconazole analogs are described in BE 841309 and U.S.P.N. 4,062,966.
  • Tolazoline (e.g., hydrochloride salt) analogs are described in U.S.P.N. 2,161,938.
  • Tolfenamic acid analogs are described in NL 6600251 and U.S.P.N. 3,313,848.
  • Triamterene analogs are described in U.S.P.N. 3,081,230.
  • Triflupromazine (e.g., hydrochloride salt) analogs are described in GB 813861.
  • Tulobuterol (e.g., hydrochloride salt) analogs are described in German patent DE 2244737.
  • Vincamine analogs are described in German patent DE 2115718 and U. S. P. N. 3,770,724.
  • Warfarin analogs are described in U.S.P.N. 2,427,578, U.S.P.N. 2,765,321, U.S.P.N.
  • Xylazine (e.g., hydrochloride salt) analogs are described in BE 634552, DE 1173475, and U.S.P.N. 3,235,550. All of these references are hereby incorporated by reference.
  • the patient may also receive additional therapeutic regimens.
  • therapeutic agents may be administered with the agent or agents described herein at concentrations known to be effective for such therapeutic agents.
  • Particularly useful agents include those that reduce glucose levels or those used to treat, prevent, or reduce metabolic disorders.
  • Such agents include those that alter the insulin signaling pathway, such as agents that increase the insulin supply, reduce insulin resistance, increase the effectiveness of insulin, reduce hepatic glucose output, control blood glucose and triglyceride levels, and reduce the absorption of carbohydrates from the intestine.
  • Exemplary agents are sulfonylureas (e.g., acetohexamide, chlorpropamide, tolazamide, tolbutamide, glimepiride, glipizide, and glyburide), non-sulfonylurea secretagogues (e.g., nateglinide and repaglinide), insulin, insulin analogs (e.g., insulin lispro, insulin aspart, insulin glarginine, NPH, lente insulin, ultralente insulin, humulin, novolin), glucagon-like peptides (e.g., GLP-I), exendin-4 (e.g., AAC2993), beta-3 adrenergic receptor agonists (e.g., YM178), PPAR gamma agonists (e.g., FK614), dipeptidyl peptidase IV inhibitors, biguanides (e.g., metformin and metformin
  • NSAIDs e.g., naproxen sodium, diclofenac sodium, diclofenac potassium, aspirin, sulindac, difhmisal, piroxicam, indomethacin, ibuprofen, nabumetone, choline magnesium trisalicylate, sodium salicylate, salicylsalicylic acid, fenoprofen, flurbiprofen, ketoprofen, meclofenamate sodium, meloxicam, oxaprozin, sulindac, and tolmetin), COX-2 inhibitors (e.g., rofecoxib, celecoxib, valdecoxib, and lumiracoxib), DMARD, anti-cytokine agents or agents that modulate the immune response to positively effect disease, such as agents that influence cell adhesion, or biologies (i.e., agents that block the action
  • therapeutic agents may be delivered separately or may be admixed into a single formulation.
  • routes of administration may be employed.
  • Routes of administration for the various embodiments include, but are not limited to, topical, transdermal, and systemic administration (such as, intravenous, intramuscular, subcutaneous, inhalation, rectal, buccal, vaginal, intraperitoneal, intraarticular, ophthalmic or oral administration).
  • systemic administration refers to all nondermal routes of administration, and specifically excludes topical and transdermal routes of administration.
  • the agent of the invention and additional therapeutic agents are administered within at least 1, 2, 4, 6, 10, 12, 18, 24 hours, 3 days, 7 days, or 14 days apart.
  • the dosage and frequency of administration of each component of the combination can be controlled independently.
  • one compound may ⁇ be administered three times per day, while the second compound may be administered once per day.
  • Combination therapy may be given in on-and-off cycles that include rest periods so that the patient's body has a chance to recover from any as yet unforeseen side effects.
  • the compounds may also be formulated together such that one administration delivers both compounds.
  • any of the agents of the combination may be administered in a low dosage or in a high dosage, each of which is defined herein.
  • the therapeutic agents of the invention may be admixed with additional active or inert ingredients, e.g., in conventional pharmaceutically acceptable carriers.
  • a pharmaceutical carrier can be any compatible, non-toxic substance suitable for the administration of the compositions of the present invention to a mammal.
  • Pharmaceutically acceptable carriers include, for example, water, saline, buffers and other compounds described for example in the Merck Index, Merck & Co., Rahway, New Jersey. Slow release formulation or a slow release apparatus may be also be used for continuous administration.
  • the second therapeutic regimen may involve transplantation of insulin-producing cells, tissues, or organs (e.g., pancreatic cells, beta pancreatic cells, or pancreas) or a modification to the lifestyle of the patient being treated.
  • Such lifestyle changes may be helpful to control glucose levels and include weight loss, physical exercise, diet control, reduction in alcohol intake, or reduction in smoking.
  • a corticosteroid may be formulated in the composition of the invention or administered to the mammal being treated according to the invention.
  • Suitable corticosteroids include 11 -alpha, 17-alpha,21-trihydroxypregn-4-ene-3,20-dione; 11 -beta, 16-alpha, 17,21 -tetrahydroxypregn-4-ene-3 ,20-dione; 11 -beta, 16- alpha, 17,21 -tetrahydroxypregn- 1 ,4-diene-3 ,20-dione; 11 -beta, 17-alpha,21 - trihydroxy-6-alpha-methylpregn-4-ene-3,20-dione; 11-dehydrocorticosterone; 11- deoxycortisol; l l-hydroxy-l,4-androstadiene-3,17-dione; 11-ketotestosterone; 14- hydroxyandrost-4-ene-3,6,17-tri
  • prednisolone metasulphobenzoate prednisolone sodium phosphate; prednisolone steaglate; prednisolone tebutate; prednisolone tetrahydrophthalate; prednisone; prednival; prednylidene; pregnenolone; procinonide; tralonide; progesterone; promegestone; rhapontisterone; rimexolone; roxibolone; rubrosterone; stizophyllin; tixocortol; topterone; triamcinolone; triamcinolone acetonide; triamcinolone acetonide 21-palmitate; triamcinolone diacetate; triamcinolone hexacetonide; trimegestone; turkesterone; and wortmannin.
  • the dosage of corticosteroid administered is a dosage equivalent to a prednisolone dosage, as defined herein.
  • a low dosage of a corticosteroid may be considered as the dosage equivalent to a low dosage of prednisolone.
  • the present invention may also involve the administration of a non- steroidal immunophilin-dependent immunosuppressant (NsIDI).
  • NsIDI non- steroidal immunophilin-dependent immunosuppressant
  • the immune system uses cellular effectors, such as B-cells and T-cells, to target infectious microbes and abnormal cell types while leaving normal cells intact.
  • an autoimmune disorder e.g., diabetes
  • Calcineurin inhibitors e.g., cyclosporines, tacrolimus, pimecrolimus
  • rapamycin target many types of immunoregulatory cells, including T-cells, and suppress the immune response in autoimmune disorders.
  • Immunosuppressants are particularly useful if the mammal is also receiving an organ, tissue, or cellular implant. Exemplary immunosuppressants are provided below.
  • the cyclosporines are fungal metabolites that comprise a class of cyclic oligopeptides that act as immunosuppressants.
  • Cyclosporine A and its deuterated analogue ISAtx247, is a hydrophobic cyclic polypeptide consisting of eleven amino acids. Cyclosporine A binds and forms a complex with the intracellular receptor cyclophilin. The cyclosporine/cyclophilin complex binds to and inhibits calcineurin, a Ca 2+ -calmodulin-dependent sei ⁇ ne-threonine-specif ⁇ c protein phosphatase.
  • Cyclosporines and their functional and structural analogs suppress the T-cell-dependent immune response by inhibiting antigen-triggered signal transduction. This inhibition decreases the expression of proinflammatory cytokines, such as IL-2.
  • Many cyclosporines e.g., cyclosporine A, B, C, D, E, F, G, H, and I
  • Cyclosporine A is a commercially available under the trade name NEORAL from Novartis.
  • Cyclosporine A structural and functional analogs include cyclosporines having one or more fluorinated amino acids (described, e.g., in U.S. Patent No. 5,227,467); cyclosporines having modified amino acids (described, e.g., in U.S. Patent Nos. 5,122,511 and 4,798,823); and deuterated cyclosporines, such as ISAtx247 (described in U.S. Patent Publication No. 20020132763). Additional cyclosporine analogs are described in U.S. Patent Nos. 6,136,357, 4,384,996, 5,284,826, and 5,709,797.
  • Cyclosporine analogs include, but are not limited to, D-Sar ( ⁇ -SMe) 3 Val 2 -DH-Cs (209-825), Allo-Thr-2-Cs, Norvaline-2-Cs, D-AIa (3-acetylamino)-8-Cs, Thr-2-Cs, and D-MeSer-3-Cs, D- Ser (0-CH 2 CH 2 -OH)- 8-Cs, and D-Ser-8-Cs, which are described in Cruz et al. (Antimicrob. Agents Chemother. 44: 143-149, 2000).
  • Cyclosporines are highly hydrophobic and readily precipitate in the presence of water (e.g., on contact with body fluids). Methods of providing cyclosporine formulations with improved bioavailability are described in U.S. Patent Nos. 4,388,307, 6,468,968, 5,051,402, 5,342,625, 5,977,066, and 6,022,852. Cyclosporine microemulsion compositions are described in U.S. Patent Nos. 5,866,159, 5,916,589, 5,962,014, 5,962,017, 6,007,840, and 6,024,978.
  • Cyclosporines can be administered either intravenously or orally, but oral administration is preferred.
  • an intravenous cyclosporine A is usually provided in an ethanol-polyoxyethylated castor oil vehicle that must be diluted prior to administration.
  • Cyclosporine A may be provided, e.g., as a microemulsion in a 25 mg or 100 mg tablets, or in a 100 mg/ml oral solution (NEORALTM).
  • patient dosage of an oral cyclosporine varies according to the patient's condition, but some standard recommended dosages in prior art treatment regimens are provided herein.
  • Patients undergoing organ transplant typically receive an initial dose of oral cyclosporine A in amounts between 12 and 15 mg/kg/day. Dosage is then gradually decreased by 5% per week until a 7-12 mg/kg/day maintenance dose is reached. For intravenous administration, 2-6 mg/kg/day is preferred for most patients. Often cyclosporines are administered in combination with other immunosuppressive agents, such as glucocorticoids. Additional information is provided in Table 3. Table 3— NsIDIs
  • Tacrolimus (PROGRAF, Fujisawa), also known as FK506, is an immunosuppressive agent that targets T-cell intracellular signal transduction pathways. Tacrolimus binds to an intracellular protein FK506 binding protein (FKBP- 12) that is not structurally related to cyclophilin (Harding et al. Nature 341:758-7601, 1989; Siekienka et al. Nature 341:755-757, 1989; and Soltoff et al., J. Biol. Chem. 267:17472-17477, 1992).
  • FKBP/FK506 complex binds to calcineurin and inhibits calcineurin's phosphatase activity.
  • NFAT a nuclear component that initiates gene transcription required for lymphokine (e.g., IL-2, gamma interferon) production and T-cell activation.
  • lymphokine e.g., IL-2, gamma interferon
  • tacrolimus inhibits T- cell activation.
  • Tacrolimus is a macrolide antibiotic that is produced by Streptomyces tsukubaensis . It suppresses the immune system and prolongs the survival of transplanted organs. It is currently available in oral and injectable formulations.
  • Tacrolimus capsules contain 0.5 mg, 1 mg, or 5 mg of anhydrous tacrolimus within a gelatin capsule shell.
  • the injectable formulation contains 5 mg anhydrous tacrolimus in castor oil and alcohol that is diluted with 9% sodium chloride or 5% dextrose prior to injection. While oral administration is preferred, patients unable to take oral capsules may receive injectable tacrolimus.
  • the initial dose should be administered no sooner than six hours after transplant by continuous intravenous infusion.
  • Tacrolimus and tacrolimus analogs are described by Tanaka et al., (J. Am. Chem. Soc, 109:5031, 1987), and in U.S. Patent Nos. 4,894,366, 4,929,611, and 4,956,352.
  • FK506-related compounds including FR-900520, FR-900523, and FR-900525, are described in U.S. Patent No. 5,254,562; O-aryl, O-alkyl, O- alkenyl, and O-alkynylmacrolides are described in U.S. Patent Nos. 5,250,678, 532,248, 5,693,648; amino O-aryl macrolides are described in U.S. Patent No.
  • alkylidene macrolides are described in U.S. Patent No. 5,284,840; N- heteroaryl, N-alkylheteroaryl, N-alkenylheteroaryl, and N-alkynylheteroaryl macrolides are described in U.S. Patent No. 5,208,241; aminomacrolides and derivatives thereof are described in U.S. Patent No. 5,208,228; fluoromacrolides are described in U.S. Patent No. 5,189,042; amino O-alkyl, O-alkenyl, and O- alkynylmacrolides are described in U.S. Patent No. 5,162,334; and halomacrolides are described in U.S. Patent No. 5,143,918.
  • While suggested dosages will vary with a patient's condition, standard recommended dosages used in prior art treatment regimens are provided below.
  • Patients having a transplanted organ typically receive doses of 0.1 -0.2 mg/kg/day of oral tacrolimus.
  • Patients being treated for rheumatoid arthritis typically receive 1-3 mg/day oral tacrolimus.
  • 0.01-0.15 mg/kg/day of oral tacrolimus is administered to a patient.
  • Patients receiving oral tacrolimus capsules typically receive the first dose no sooner than six hours after transplant, or eight to twelve hours after intravenous tacrolimus infusion was discontinued.
  • suggested tacrolimus dosages include 0.005-0.01 mg/kg/day, 0.01-0.03 mg/kg/day, 0.03-0.05 mg/kg/day, 0.05-0.07 mg/kg/day, 0.07-0.10 mg/kg/day, 0.10-0.25 mg/kg/day, or 0.25-0.5 mg/kg/day.
  • Tacrolimus is extensively metabolized by the mixed- function oxidase system, in particular, by the cytochrome P-450 system.
  • the primary mechanism of metabolism is demethylation and hydroxylation. While various tacrolimus metabolites are likely to exhibit immunosuppressive biological activity, the 13- demethyl metabolite is reported to have the same activity as tacrolimus.
  • Ascomycin is a close structural analog of FK506 and is a potent immunosuppressant. It binds to FKBP- 12 and suppresses its proline rotamase activity.
  • the ascomycin-FKBP complex inhibits calcineurin, a type 2B phosphatase.
  • Pimecrolimus (also known as SDZ ASM-981) is an 33-epi-chloro derivative of the ascomycin. It is produced by the strain Streptomyces hygroscopicus var. as corny DCtus. Like tacrolimus, pimecrolimus (ELIDELTM, Novartis) binds FKBP- 12, inhibits calcineurin phosphatase activity, and inhibits T-cell activation by blocking the transcription of early cytokines. In particular, pimecrolimus inhibits IL-2 production and the release of other proinflammatory cytokines. Pimecrolimus structural and functional analogs are described in U.S. Patent
  • Pimecrolimus is particularly useful for the treatment of atopic dermatitis. Pimecrolimus is currently available as a 1% cream. While individual dosing will vary with the patient's condition, some standard recommended dosages are provided below. Patients having an organ transplant can be administered 160-240 mg/day of pimecrolimus. Thus, useful dosages of pimecrolimus range between 0.5-5 mg/day, between 5-10 mg/day, between 10-30 mg/day, between 40-80 mg/day, between 80-120 mg/day, or even between 120- 200 mg/day.
  • Rapamycin (Rapamune® sirolimus, Wyeth) is a cyclic lactone produced by Steptomyces hygroscopicus. Rapamycin is an immunosuppressive agent that inhibits T-lymphocyte activation and proliferation. Like cyclosporines, tacrolimus, and pimecrolimus, rapamycin forms a complex with the immunophilin FKBP-12, but the rapamycin-FKBP-12 complex does not inhibit calcineurin phosphatase activity. The rapamycin-immunophilin complex binds to and inhibits the mammalian target of rapamycin (mTOR), a kinase that is required for cell cycle progression. Inhibition of mTOR kinase activity blocks T-lymphocyte proliferation and lymphokine secretion.
  • mTOR mammalian target of rapamycin
  • Rapamycin structural and functional analogs include mono- and diacylated rapamycin derivatives (U.S. Patent No. 4,316,885); rapamycin water-soluble prodrugs (U.S. Patent No. 4,650,803); carboxylic acid esters (PCT Publication No. WO 92/05179); carbamates (U.S. Patent No. 5,118,678); amide esters (U.S. Patent No. 5,118,678); biotin esters (U.S. Patent No. 5,504,091); fluorinated esters (U.S. Patent No. 5,100,883); acetals (U.S. Patent No. 5,151,413); silyl ethers (U.S. Patent No.
  • Everolimus 40-O-(2-hydroxyethyl)rapamycin; CERTICANTM; Novartis
  • CERTICANTM is an immunosuppressive macrolide that is structurally related to rapamycin, and has been found to be particularly effective at preventing acute rejection of organ transplant when given in combination with cyclosporin A.
  • Rapamycin is currently available for oral administration in liquid and tablet formulations.
  • RAPAMUNETM liquid contains 1 mg/mL rapamycin that is diluted in water or orange juice prior to administration. Tablets containing 1 or 2 mg of rapamycin are also available. Rapamycin is preferably given once daily as soon as possible after transplantation. It is absorbed rapidly and completely after oral administration.
  • patient dosage of rapamycin varies according to the patient's condition, but some standard recommended dosages are provided below.
  • the initial loading dose for rapamycin is 6 mg. Subsequent maintenance doses of 2 mg/day are typical.
  • a loading dose of 3 mg, 5 mg, 10 mg, 15 mg, 20 mg, or 25 mg can be used with a 1 mg, 3 mg, 5 mg, 7 mg, or 10 mg per day maintenance dose.
  • rapamycin dosages are typically adjusted based on body surface area; generally a 3 mg/m 2 /day loading dose and a l-mg/m 2 /day maintenance dose is used.
  • the drugs used in any of the combinations described herein may be covalently attached to one another to form a conjugate of formula XXX.
  • (A) is a drug listed on Table 1 covalently tethered via a linker (L) to (B), a sulfonylurea, a non-sulfonylurea secretagogue, insulin, an insulin analog, glucagon-like peptide, exendin-4, YMl 78, FK614, a dipeptidyl peptidase IV inhibitor, biguanide, thiazalidinedione, an alpha-glucosidase inhibitor, an immunosuppressant, an immmunomodulator, an angiotensin converting enzyme (ACE) inhibitor, an angiotensin II receptor blocker, an antioxidant, or a second drug listed on Table 1.
  • ACE angiotensin converting enzyme
  • Conjugates of the invention can be administered to a subject by any route and for the treatment of any disease described herein.
  • the conjugates of the invention can be prodrugs, releasing drug (A) and drug (B) upon, for example, cleavage of the conjugate by intracellular and extracellular enzymes (e.g., amidases, esterases, and phosphatases).
  • the conjugates of the invention can also be designed to largely remain intact in vivo, resisting cleavage by intracellular and extracellular enzymes. The degradation of the conjugate in vivo can be controlled by the design of linker (L) and the covalent bonds formed with drug (A) and drug (B) during the synthesis of the conjugate.
  • Conjugates can be prepared using techniques familiar to those skilled in the art.
  • the conjugates can be prepared using the methods disclosed in G. Hermanson, Bioconjugate Techniques, Academic Press, Inc., 1996.
  • the synthesis of conjugates may involve the selective protection and deprotection of alcohols, amines, ketones, sulfhydryls or carboxyl functional groups of drug (A), the linker, and/or drug (B).
  • commonly used protecting groups for amines include carbamates, such as tert-butyl, benzyl, 2,2,2-trichloroethyl, 2- trimethylsilylethyl, 9-fluorenylmethyl, allyl, and m-nitrophenyl.
  • amides such as foraiamides, acetamides, trifluoroacetamides, sulfonamides, trifluoromethanesulfonyl amides, trimethylsilylethanesulfonamides, and tert-butylsulfonyl amides.
  • protecting groups for carboxyls include esters, such as methyl, ethyl, tert-butyl, 9-fluorenylmethyl, 2-(trimethylsilyl)ethoxy methyl, benzyl, diphenylmethyl, O-nitrobenzyl, ortho-esters, and halo-esters.
  • Examples of commonly used protecting groups for alcohols include ethers, such as methyl, methoxymethyl, methoxyethoxymethyl, methylthiomethyl, benzyloxymethyl, tetrahydropyranyl, ethoxyethyl, benzyl, 2-napthylmethyl, O-nitrobenzyl, P- nitrobenzyl, P-methoxybenzyl, 9-phenylxanthyl, trityl (including methoxy-trityls), and silyl ethers.
  • Examples of commonly used protecting groups for sulfhydryls include many of the same protecting groups used for hydroxyls.
  • sulfhydryls can be protected in a reduced form (e.g., as disulfides) or an oxidized form (e.g., as sulfonic acids, sulfonic esters, or sulfonic amides).
  • Protecting groups can be chosen such that selective conditions (e.g., acidic conditions, basic conditions, catalysis by a nucleophile, catalysis by a lewis acid, or hydrogenation) are required to remove each, exclusive of other protecting groups in a molecule.
  • the conditions required for the addition of protecting groups to amine, alcohol, sulfhydryl, and carboxyl functionalities and the conditions required for their removal are provided in detail in T. W. Green and P.G.M. Wuts, Protective Groups in Organic Synthesis (2 nd Ed.), John Wiley & Sons, 1991 and PJ. Kocienski, Protecting Groups, Georg Thieme Verlag, 1994. Additional synthetic details are provided below. Linkers
  • the linker component of the invention is, at its simplest, a bond between drug (A) and drug (B), but typically provides a linear, cyclic, or branched molecular skeleton having pendant groups covalently linking drug (A) to drag (B).
  • linking of drug (A) to drug (B) is achieved by covalent means, involving bond formation with one or more functional groups located on drug (A) and drag (B).
  • Examples of chemically reactive functional groups which may be employed for this purpose include, without limitation, amino, hydroxyl, sulfhydryl, carboxyl, carbonyl, carbohydrate groups, vicinal diols, thioethers, 2- aminoalcohols, 2-aminothiols, guanidinyl, imidazolyl, and phenolic groups.
  • the covalent linking of drag (A) and drag (B) may be effected using a linker which contains reactive moieties capable of reaction with such functional groups present in drag (A) and drag (B).
  • a linker which contains reactive moieties capable of reaction with such functional groups present in drag (A) and drag (B).
  • an amine group of drag (A) may react with a carboxyl group of the linker, or an activated derivative thereof, resulting in the formation of an amide linking the two.
  • N-Maleimide derivatives are also considered selective towards sulfhydryl groups, but may additionally be useful in coupling to amino groups under certain conditions.
  • Reagents such as 2-iminothiolane (Traut et al., Biochemistry 12:3266 (1973)), which introduce a thiol group through conversion of an amino group, may be considered as sulfhydryl reagents if linking occurs through the formation of disulphide bridges.
  • Examples of reactive moieties capable of reaction with amino groups include, for example, alkylating and acylating agents.
  • N-maleimide derivatives which may react with amino groups either through a Michael type reaction or through acylation by addition to the ring carbonyl group, for example, as described by Smyth et al., J. Am. Chem. Soc. 82:4600 (1960) wd Biochem. J. 91:589 (1964);
  • aryl halides such as reactive nitrohaloaromatic compounds
  • alkyl halides as described, for example, by McKenzie et al., J. Protein Chem. 7:581 (1988);
  • epoxide derivatives such as epichlorohydrin and bisoxiranes, which may react with amino, sulfhydryl, or phenolic hydroxyl groups;
  • (x) ⁇ -haloalkyl ethers which are more reactive alkylating agents than normal alkyl halides because of the activation caused by the ether oxygen atom, as described by Benneche et al., Eur. J. Med. Chem. 28:463 (1993).
  • Representative amino-reactive acylating agents include: (i) isocyanates and isothiocyanates, particularly aromatic derivatives, which form stable urea and thiourea derivatives respectively; (ii) sulfonyl chlorides, which have been described by Herzig et al., Biopolymers 2:349 (1964); (iii) acid halides;
  • active esters such as nitrophenylesters or N-hydroxysuccinimidyl esters
  • Aldehydes and ketones may be reacted with amines to form Schiff s bases, which may advantageously be stabilized through reductive animation.
  • Alkoxylamino moieties readily react with ketones and aldehydes to produce stable alkoxamines, for example, as described by Webb et al., in Bioconjugate Chem. 1 :96 (1990).
  • reactive moieties capable of reaction with carboxyl groups include diazo compounds such as diazoacetate esters and diazoacetamides, which react with high specificity to generate ester groups, for example, as described by Herriot, Adv. Protein Chem. 3:169 (1947).
  • Carboxyl modifying reagents such as carbodiimides, which react through O-acylurea formation followed by amide bond formation, may also be employed.
  • functional groups in drug (A) and/or drug (B) may, if desired, be converted to other functional groups prior to reaction, for example, to confer additional reactivity or selectivity.
  • methods useful for this purpose include conversion of amines to carboxyls using reagents such as dicarboxylic anhydrides; conversion of amines to thiols using reagents such as N-acetylhomocysteine thiolactone, S-acetylmercaptosuccinic anhydride, 2- iminothiolane, or thiol- containing succinimidyl derivatives; conversion of thiols to carboxyls using reagents such as ⁇ -haloacetates; conversion of thiols to amines using reagents such as ethylenimine or 2-bromoethylamine; conversion of carboxyls to amines using reagents such as carbodiimides followed by diamines; and conversion of alcohol
  • So-called zero-length linkers involving direct covalent joining of a reactive chemical group of drag (A) with a reactive chemical group of drag (B) without introducing additional linking material may, if desired, be used in accordance with the invention.
  • the linker will include two or more reactive moieties, as described above, connected by a spacer element.
  • the presence of such a spacer permits bifunctional linkers to react with specific functional groups within drag (A) and drug (B), resulting in a covalent linkage between the two.
  • the reactive moieties in a linker may be the same (homobifunctional linker) or different (heterobifunctional linker, or, where several dissimilar reactive moieties are present, heteromultifunctional linker), providing a diversity of potential reagents that may bring about covalent attachment between drag (A) and drag (B).
  • Spacer elements in the linker typically consist of linear or branched chains and may include a C ⁇ 10 alkyl, C 2 _ 10 alkenyl, C 2 _i 0 alkynyl, C 2 _ 6 heterocyclyl, C 6 _ 12 aryl, C 7 _ J4 alkaryl, C 3 _i 0 alkheterocyclyl, or C 1 ⁇ o heteroalkyl.
  • the linker is described by formula (XXXI):
  • G 1 is a bond between drug (A) and the linker;
  • G 2 is a bond between the linker and drug (B);
  • Z 1 , Z 2 , Z 3 , and Z 4 each, independently, is selected from O, S, and NR 31 ;
  • R 31 is hydrogen, Ci_ 4 alkyl, C 2 _ 4 alkenyl, C 2 _ ⁇ alkynyl, C 2 _ 6 heterocyclyl, C 6 _i 2 aryl, C 7 _ 14 alkaryl, C 3 _ 10 alkheterocyclyl, or Ci_ 7 heteroalkyl;
  • Y 1 and Y 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl;
  • o, p, s, t, u, and v are each, independently, 0 or 1;
  • R 3 o is a C 1 ⁇ o alkyl, C 2 ⁇ o alkenyl, C 2
  • homobifunctional linkers useful in the preparation of conjugates of the invention include, without limitation, diamines and diols selected from ethylenediamine, propylenediamine and hexamethylenediamine, ethylene glycol, diethylene glycol, propylene glycol, 1 ,4-butanediol, 1 ,6-hexanediol, cyclohexanediol, and polycaprolactone diol.
  • any of the agents employed according to the present invention may be contained in any appropriate amount in any suitable carrier substance, and is generally present in an amount of 1-95% by weight of the total weight of the composition.
  • the composition may be provided in a dosage form that is suitable for the oral, parenteral (e.g., intravenously, intramuscularly), rectal, cutaneous, nasal, vaginal, inhalant, skin (patch), or ocular administration route.
  • the composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, or aerosols.
  • the pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy, 20th edition, 2000, ed. A.R. Gennaro, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
  • each agent may be formulated in a variety of ways that are known in the art. Desirably, the agents are formulated together for the simultaneous or near simultaneous administration of the agents. Such co-formulated compositions can include the two agents formulated together in the same pill, capsule, liquid, etc. It is to be understood that, when referring to the formulation of such combinations, the formulation technology employed is also useful for the formulation of the individual agents of the combination, as well as other combinations of the invention. By using different formulation strategies for different agents, the pharmacokinetic profiles for each agent can be suitably matched.
  • kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc.
  • the kit can include optional components that aid in the administration of the unit dose to patients, such as vials for reconstituting powder forms, syringes for injection, customized IV delivery systems, inhalers, etc.
  • the unit dose kit can contain instructions for preparation and administration of the compositions.
  • the kit may be manufactured as a single use unit dose for one patient, multiple uses for a particular patient (at a constant dose or in which the individual compounds may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple patients ("bulk packaging").
  • the kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.
  • the dosage of any of the agents of the combination of the invention will depend on the nature of the agent, and can readily be determined by one skilled in the art. Typically, such dosage is normally about 0.001 mg to 2000 mg per day, desirably about 1 mg to 1000 mg per day, and more desirably about 5 mg to 500 mg per day. Dosages up to 200 mg per day may be necessary.
  • Administration of each drug in the combination can, independently, be one to four times daily for one day to one year, and may even be for the life of the patient. Chronic, long-term administration will be indicated in many cases.
  • the compounds of the invention may be employed in mechanistic assays to determine whether other combinations, or single agents, are as effective as the combination in treating, reducing, or preventing metabolic disorders (e.g., diabetes or any of its associated conditions) using assays generally known in the art, examples of which are described herein.
  • candidate compounds may be tested, alone or in combination (e.g., with an agent that reduces glucose levels, such as those described herein) and applied to adipocytes or muscle cells in the presence of insulin and glucose. After a suitable time, these cells are examined for glucose uptake. An increase in glucose uptake identifies a candidate compound or combination of agents as an effective agent to treat, prevent, or reduce a metabolic disorder.
  • the agents of the invention are also useful tools in elucidating mechanistic information about the biological pathways involved in glucose transport and glucose utilization. Such information can lead to the development of new combinations or single agents for treating, preventing, or reducing diabetes.
  • Methods known in the art to determine biological pathways can be used to determine the pathway, or network of pathways affected by contacting cells (e.g., adipocytes, muscle cells, or any cells that utilizes glucose as a source of energy) in the presence of insulin and glucose or by contacting pancreatic cells (any cell that has the ability to produce insulin) with the compounds of the invention.
  • Such methods can include, analyzing cellular constituents that are expressed or repressed after contact with the compounds of the invention as compared to untreated, positive or negative control compounds, and/or new single agents and combinations, or analyzing some other metabolic activity of the cell such as enzyme activity, nutrient uptake, and proliferation.
  • Cellular components analyzed can include gene transcripts, and protein expression.
  • Suitable methods can include standard biochemistry techniques, radiolabeling the compounds of the invention (e.g., 1 C or 3 H labeling), and observing the compounds binding to proteins, e.g. using 2D gels, gene expression profiling. Once identified, such compounds can be used in in vivo models (e.g., NOD mice) to further validate the tool or develop new agents or strategies to treat, prevent, or reduce metabolic disorders.
  • the methods of this invention may also be used prophylactically, in patients who are an increased risk of developing diabetes or a condition associated with diabetes.
  • Risk factors include for example, family history of diabetes or obesity conditions, quality of nutrition, level of physical activity, presence of molecular markers of diabetes, age, race, or sex. Patients affected with other non-related disorders may also be predisposed to secondary diabetes.
  • Exemplary candidate compounds Peptide moieties include for example, family history of diabetes or obesity conditions, quality of nutrition, level of physical activity, presence of molecular markers of diabetes, age, race, or sex. Patients affected with other non-related disorders may also be predisposed to secondary diabetes.
  • Exemplary candidate compounds Peptide moieties include for example, family history of diabetes or obesity conditions, quality of nutrition, level of physical activity, presence of molecular markers of diabetes, age, race, or sex. Patients affected with other non-related disorders may also be predisposed to secondary diabetes.
  • Exemplary candidate compounds Peptide moieties include for example, family history of diabetes or obesity conditions, quality of
  • Peptides, peptide mimetics, and peptide fragments are suitable for use in practicing the invention.
  • exemplary inhibitors include compounds that reduce the amount of target protein or RNA levels (e.g., antisense compounds, dsRNA, ribozymes) and compounds that compete with endogenous mitotic kinesins or protein tyrosine phosphatases for binding partners (e.g., dominant negative proteins or polynucleotides encoding the same).
  • RNA secondary structure folding program such as MFOLD (M. Zuker, D. H. Mathews & D. H. Turner, Algorithms and Thermodynamics for RNA Secondary Structure Prediction: A Practical Guide. In: RNA Biochemistry and Biotechnology, J. Barciszewski & B. F. C. Clark, eds., NATO ASI Series, Kluwer Academic Publishers, (1999)).
  • Sub-optimal folds with a free energy value within 5% of the predicted most stable fold of the mRNA are predicted using a window of 200 bases within which a residue can find a complimentary base to form a base pair bond. Open regions that do not form a base pair are summed together with each suboptimal fold and areas that are predicted as open are considered more accessible to the binding to antisense nucleobase oligomers.
  • Other methods for antisense design are described, for example, in U.S. Patent No. 6,472,521, Antisense Nucleic Acid Drug Dev. 1997 7:439-444, Nucleic Acids Research 28:2597-2604, 2000, and Nucleic Acids Research 31:4989-4994, 2003.
  • RNA interference The biological activity of a signaling molecule can be reduced through the use of RNA interference (RNAi), employing, e.g., a double stranded RNA (dsRNA) or small interfering RNA (siRNA) directed to the signaling molecule in question (see, e.g., Miyamoto et al., Prog. Cell Cycle Res. 5:349-360, 2003; U.S. Patent Application Publication No. 20030157030).
  • dsRNA double stranded RNA
  • siRNA small interfering RNA
  • Methods for designing such interfering RNAs are known in the art. For example, software for designing interfering RNA is available from Oligoengine (Seattle, WA).
  • Example 1 An increase in insulin-stimulated glucose uptake in vitro
  • mice adipocytes were employed to identify combinations of agents that have the ability to increase glucose uptake upon insulin stimulation, as detected by scintillation counting of radiolabeled glucose (using, for example, the Perkin Elmer 1450 Microbeta JET reader). These assays were conducted as follows. Materials and methods Prees media
  • Prees media Complete media, also referred to as "Prees” media, was prepared as follows. Dulbecco's Modified Eagle's Medium (DMEM) was supplemented with L-glutamine, penicillin-G and streptomycin (pen/strep), and heat-inactivated fetal bovine serum (FBS) (heat inactivated at 65 0 C for 30 minutes). Because serum can affect the growth, adherence, and differentiation of cells, any new lot of serum was first tested prior to use. Media was equilibrated in the incubator (5% CO 2 ) until the pH was within the proper range ( ⁇ 7), as indicated by the red/orange color of the indicator dye. If the media became pink (indicating a high pH), we discarded the media as basic conditions can affect cells and denature the insulin used in the differentiation medium- 1 (DMl) and the differentiation medium-2 (DM2).
  • DMEM Dulbecco's Modified Eagle's Medium
  • pen/strep penicillin-G and streptomycin
  • FBS heat
  • Differentiation media- 1 was prepared by supplementing DMEM with 10% FBS, L-glutamine, pen/strep, IBMX (375 ⁇ M), insulin (120 nM), and dexamethasone (188 nM).
  • Differentiation media-2 was prepared by supplementing DMEM with 10% FBS, L-glutamine, pen/strep, and insulin (120 nM).
  • Cell culture plates were gelatinized as follows. Gelatin (l%w/v in distilled water) was autoclaved and stored at room temperature. The bottom of each cell culture well was covered uniformly in the gelatin solution, ensuring that no bubbles were formed. This solution was removed leaving behind a thin film of gelatin. These plates were left to dry under the tissue culture hood. Plates were next washed with PBS, after which a 0.5% glutaric dialdehyde solution (glutaraldehyde in distilled water) was added to the cell culture wells. After ten minutes, wells were washed twice with DMEM containing pen-strep. Each washing step should last for approximately five minutes.
  • D-(+)-glucose (“cold" glucose, not radiolabeled) was added to DPBS mix to a final concentration of 1O mM.
  • Lysis buffer a mixture of a base (e.g., sodium hydroxide at a final concentration of 0.5N) and a detergent (e.g., sodium dodecyl sulphate (SDS) diluted to a final concentration of 0.1% w/v) was freshly prepared each time (within one to two hours of use). Prior to use, lysis buffer was warmed up to a temperature exceeding that of room temperature for a period of approximately 30 minutes to avoid precipitation of the buffer.
  • a base e.g., sodium hydroxide at a final concentration of 0.5N
  • a detergent e.g., sodium dodecyl sulphate (SDS) diluted to a final concentration of 0.1% w/v
  • Pre-adipocyte 3T3-L1 cells were plated at a density of approximately 5000 cells/well (in black NUNC 96 well plate). These cells were differentiated into adipocytes in two separate steps. Initially, cells were cultured in differentiation medium- 1 (DMl) (day 1 of adipocyte differentation) for a period of two to three days. DMl prevents proliferation and induces the expression of adipocyte- specific genes. Cells were next cultured in differentiation medium-2 (DM2) for 3 to 4 days, after which the culture media was replaced by fresh DM2. The glucose uptake assay was performed at day 9-15 of differentiation.
  • DMl differentiation medium- 1
  • DM2 differentiation medium-2
  • DM2 was removed and replaced with fresh Prees media.
  • Candidate compounds were added at this time, allowing an incubation period of approximately 48 hours.
  • cells now at day 9 to 15 of differentiation
  • DPBS magnesium sulfate
  • Hepes (1OmM) at pH ⁇ 7.
  • fresh DPBS containing insulin (10 iiM) was added to the adipocytes.
  • Fresh DPBS without any insulin were placed on cells that served as a negative control.
  • radioactive glucose (labeled with 14 C , at a final concentration of 0.04 mM, ⁇ 0.26 ⁇ Ci 14 C-glucose in each well) was added to the media for a period of 15 minutes at room temperature. Media was next removed and cells were washed thoroughly and lysed. Upon lysis, cells form a small, cloudy mass, detached from the well bottom. 10% glacial acetic acid was added to each well to neutralize the lysis reaction. Scintillation fluid was next added to the wells and the incorporation of glucose was determined by measuring the amount of radioactivity in each well using the MicroBeta plate reader.
  • the average fold increase in insulin-stimulated glucose uptake compared to vehicle treated control was determined. The average of three biological replicates is shown. At all combination doses there is an increase in glucose uptake as compared to the single agents.
  • Example 2 The combination of bezafibrate and diflunisal reduce insulin sensitivity in a rat model Insulin resistance was induced in male Sprague Dawley rats by four weeks of high fat feeding (60% of calories derived from fat). Drug treatment began one week after initiation of high fat diet. Drugs were administered daily, by oral gavage for a three week period.
  • HOMA homeostasis model assessment
  • HOMA fasting serum glucose X fasting serum insulin
  • Example 3 Screening assays identify additional agents that increase glucose uptake by adipocytes
  • Differentiated mouse adipocytes were employed to identify combinations of agents that have the ability to increase glucose uptake upon insulin stimulation, as detected by scintillation counting of radiolabeled glucose (using, for example, the Perkin Elmer 1450 Microbeta JET reader). These assays were conducted as follows.
  • Prees media Complete media, also referred to as "Prees” media, was prepared as follows. Dulbecco's Modified Eagle's Medium (DMEM) was supplemented with L-glutamine, penicillin-G and streptomycin (pen/strep), and heat-inactivated fetal bovine serum (FBS) (heat inactivated at 65 0 C for 30 minutes). Because serum can affect the growth, adherence, and differentiation of cells, any new lot of serum was first tested prior to use. Media was equilibrated in the incubator (5% CO 2 ) until the pH was within the proper range ( ⁇ 7), as indicated by the red/orange color of the indicator dye. If the media became pink (indicating a high pH), we discarded the media as basic conditions can affect cells and denature the insulin used in the differentiation medium- 1 (DMl) and the differentiation medium-2 (DM2).
  • DMEM Dulbecco's Modified Eagle's Medium
  • pen/strep penicillin-G and streptomycin
  • FBS heat
  • Differentiation media- 1 was prepared by supplementing DMEM with 10% FBS, L-glutamine, pen/strep, IBMX (375 ⁇ M), insulin (120 iiM), and dexamethasone (188 nM).
  • Differentiation media-2 was prepared by supplementing DMEM with 10% FBS, L-glutamine, pen/strep, and insulin (120 nM).
  • Cell culture plates were gelatinized as follows. Gelatin (l%w/v in distilled water) was autoclaved and stored at room temperature. The bottom of each cell culture well was covered uniformly in the gelatin solution, ensuring that no bubbles were formed. This solution was removed leaving behind a thin film of gelatin. These plates were left to dry under the tissue culture hood. Plates were next washed with PBS, after which a 0.5% glutaric dialdehyde solution (glutaraldehyde in distilled water) was added to the cell culture wells. After ten minutes, wells were washed twice with DMEM containing pen-strep. Each washing step should last for approximately five minutes.
  • 3T3-L1 pre-adipocyte cells were split approximately every 2-3 days or upon reaching a confluence of approximately 60%. Overconfluency may affect the ability of these cells to differentiate into adipocytes.
  • D-(+)-glucose (“cold" glucose, not radiolabeled) was added to DPBS mix to a final concentration of 10 mM.
  • Lysis buffer a mixture of a base (e.g., sodium hydroxide at a final concentration of 0.5N) and a detergent (e.g., sodium dodecyl sulphate (SDS) diluted to a final concentration of 0.1% w/v) was freshly prepared each time (within one to two hours of use). Prior to use, lysis buffer was warmed up to a temperature exceeding that of room temperature for a period of approximately 30 minutes to avoid precipitation of the buffer. Determination of glucose uptake
  • Pre-adipocyte 3T3-L1 cells were plated at a density of approximately 5000 cells/well (in black NUNC 96 well plate). These cells were differentiated into adipocytes in two separate steps. Initially, cells were cultured in differentiation medium- 1 (DMl) (day 1 of adipocyte differentation) for a period of two to three days. DMl prevents proliferation and induces the expression of adipocyte- specif ⁇ c genes. Cells were next cultured in differentiation medium-2 (DM2) for 3 to 4 days, after which the culture media was replaced by fresh DM2. The glucose uptake assay was performed at day 9-15 of differentiation. Two days prior to the experiment (at day 7-13 of differentiation), DM2 was removed and replaced with fresh Prees media.
  • DMl differentiation medium- 1
  • DM2 differentiation medium-2
  • Candidate compounds were added at this time, allowing an incubation period of approximately 48 hours.
  • cells now at day 9 to 15 of differentiation
  • DPBS magnesium sulfate
  • Hepes (1OmM) at pH ⁇ 7.
  • fresh DPBS containing insulin (10 liM) was added to the adipocytes.
  • Fresh DPBS without any insulin were placed on cells that served as a negative control.
  • radioactive glucose (labeled with 14 C , at a final concentration of 0.04 niM, -0.26 ⁇ Ci 14 C-glucose in each well) was added to the media for a period of 15 minutes at room temperature. Media was next removed and cells were washed thoroughly and lysed. Upon lysis, cells form a small, cloudy mass, detached from the well bottom. 10% glacial acetic acid was added to each well to neutralize the lysis reaction. Scintillation fluid was next added to the wells and the incorporation of glucose was determined by measuring the amount of radioactivity in each well using the MicroBeta plate reader. Compounds that exhibited glucose uptake activity are shown in Table 4.
  • Table 4 shows glucose uptake activity (denoted as F (fold over base) with the standard deviation (sF)) of each of the compounds at various concentrations. S/N denotes the signal over noise ratio.
  • human skeletal myoblasts obtained by the conditional immortalization of cells derived from a non-diabetic subject, were used to screen the effect of a multitude of compounds on glycogen synthesis. For each compound, two doses were tested in triplicate. The vehicle was used as a negative control and insulin was used as a positive control. Prior to treatment, cells were serum-starved for 12 to 18 hours in Ham's FlO medium.

Abstract

L'invention concerne des compositions, des procédés, et des kits pour le traitement de troubles métaboliques de type diabète et obésité.
EP05768186A 2004-06-30 2005-06-29 Procedes et reactifs pour le traitement de troubles metaboliques Withdrawn EP1781303A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US58438004P 2004-06-30 2004-06-30
US64932905P 2005-02-02 2005-02-02
PCT/US2005/023030 WO2006004803A1 (fr) 2004-06-30 2005-06-29 Procedes et reactifs pour le traitement de troubles metaboliques

Publications (2)

Publication Number Publication Date
EP1781303A1 true EP1781303A1 (fr) 2007-05-09
EP1781303A4 EP1781303A4 (fr) 2008-07-02

Family

ID=35783202

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05768186A Withdrawn EP1781303A4 (fr) 2004-06-30 2005-06-29 Procedes et reactifs pour le traitement de troubles metaboliques

Country Status (14)

Country Link
US (1) US7795310B2 (fr)
EP (1) EP1781303A4 (fr)
JP (1) JP2008505176A (fr)
KR (1) KR20070027747A (fr)
AR (1) AR049955A1 (fr)
AU (1) AU2005259864A1 (fr)
BR (1) BRPI0512856A (fr)
CA (1) CA2571683A1 (fr)
IL (1) IL180251A0 (fr)
MX (1) MX2007000142A (fr)
NO (1) NO20070510L (fr)
RU (1) RU2007103178A (fr)
TW (1) TW200608961A (fr)
WO (1) WO2006004803A1 (fr)

Families Citing this family (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19853487A1 (de) * 1998-11-19 2000-05-25 Fumapharm Ag Muri Verwendung von Dialkylfumaraten
WO2005087234A1 (fr) 2004-03-05 2005-09-22 The Trustees Of The University Of Pennsylvania Traitement a faibles effets secondaires contre des affections liees a l'hyperlipidemie et l'hypercholesterolemie
ATE410431T1 (de) * 2004-04-14 2008-10-15 Wyeth Corp Verfahren zur herstellung von rapamycin-42-estern und fk-506-32-estern mit dicarbonsäure, vorstufen für rapamycinkonjugate und antikörper
US20070093468A1 (en) * 2005-10-18 2007-04-26 Wisler Gerald L Methods for treating disorders associated with hyperlipidemia in a mammal
FR2904935A1 (fr) * 2006-08-18 2008-02-22 Centre Nat Rech Scient Composition antidiabetique apte a stimuler la secretion d'insuline et destinee au traitement du diabete de type 2 (diabete non insulino-dependant).
EP2120927A1 (fr) * 2006-12-21 2009-11-25 Aegerion Pharmaceuticals, Inc. Procédés de traitement de l'obésité à l'aide d'une combinaison comprenant un inhibiteur de mtp et un inhibiteur de l'absorption du cholestérol
BRPI0806774A2 (pt) * 2007-01-16 2011-09-13 Ipintl Llc composição para tratamento de sìndrome metabólica
EP2120959A4 (fr) * 2007-01-23 2010-05-26 Reddys Lab Ltd Dr Procédés et compositions destinés au traitement de la résistance à l'insuline, du diabète et d'une dyslipidémie associée au diabète
CA2687715A1 (fr) * 2007-05-09 2008-11-20 Traffick Therapeutics Inc. Dosage de criblage pour identifier des correcteurs de defauts de trafic des proteines
WO2009052376A1 (fr) 2007-10-18 2009-04-23 Musc Foundation For Research Development Procédés de diagnostic d'un cancer génito-urinaire
CA2703260A1 (fr) * 2007-11-07 2009-05-14 Burnham Institute For Medical Research Procede et composes pour la regulation de la production d'insuline
BRPI0912716A2 (pt) * 2008-05-13 2015-10-13 Genmedica Therapeutics Sl composto.
CA2666036C (fr) * 2008-05-16 2017-09-12 Chien-Hung Chen Compositions inedites et methodes de traitement des maladies hyperproliferantes
US8729097B2 (en) * 2008-10-06 2014-05-20 The Johns Hopkins University Quinoline compounds as inhibitors of angiogenesis, human methionine aminopeptidase, and SIRT1, and methods of treating disorders
US8642650B2 (en) 2008-12-04 2014-02-04 Zafgen, Inc. Methods of treating an overweight or obese subject
WO2013055385A2 (fr) 2011-10-03 2013-04-18 Zafgen Corporation Méthodes de traitement de troubles liés à l'âge
WO2010065877A2 (fr) 2008-12-04 2010-06-10 Zafgen Corporation Procédés de traitement d'un sujet en surpoids ou obèse
BRPI1011593A2 (pt) * 2009-03-16 2016-03-22 Genmedica Therapeutics Sl "combinação farmacêutica, composição farmaceuticamente aceitavél, e, método para tratar um distúrbio metabólico, para tratar doença pulmonar obstrutiva crônica, e para tratar ou prevenir um ou mais distúrbios metabólicos."
WO2010106082A1 (fr) * 2009-03-16 2010-09-23 Genmedica Therapeutics Sl Conjugués anti-inflammatoires et anti-oxydants utiles pour traiter des troubles métaboliques
WO2010107702A1 (fr) * 2009-03-16 2010-09-23 Ipintl, Llc Traitement de la maladie d'alzheimer et de l'ostéoporose, et réduction du vieillissement
CN102639494B (zh) 2009-10-09 2016-11-09 扎夫根公司 砜化合物及其制备和使用方法
WO2011085198A1 (fr) 2010-01-08 2011-07-14 Zafgen Corporation Inhibiteur de metap-2 pour utilisation dans le traitement d'une hypertrophie bénigne de la prostate (bph)
CN102791699B (zh) 2010-01-08 2016-04-06 扎夫根公司 烟曲霉醇型化合物和其制造和使用方法
US20130266578A1 (en) 2010-04-07 2013-10-10 Thomas E. Hughes Methods of treating an overweight subject
WO2011150338A1 (fr) * 2010-05-27 2011-12-01 Zafgen Corporation Procédés de traitement de l'obésité
BR112013001613A2 (pt) 2010-07-22 2016-05-24 Zafgen Inc compostos tricíclicos e métodos para fazer e usar os mesmos.
JP2013543899A (ja) 2010-11-29 2013-12-09 ザフゲン,インコーポレイテッド 6−o−(4−ジメチルアミノエトキシ)シンナモイルフマギロールの非連日投与を用いた肥満の治療
US8466197B2 (en) 2010-12-14 2013-06-18 Genmedica Therapeutics Sl Thiocarbonates as anti-inflammatory and antioxidant compounds useful for treating metabolic disorders
EP2668169B1 (fr) 2011-01-26 2017-11-15 Zafgen, Inc. Composés de tétrazole et procédés de préparation et d'utilisation de ceux-ci
WO2012103520A1 (fr) * 2011-01-28 2012-08-02 Board Of Regents Of The University Of Nebraska Procédés et compositions pour moduler la cyclophiline d
CA2829204C (fr) 2011-03-08 2019-12-24 Zafgen, Inc. Derives d'oxaspiro[2.5]octane et analogues
AU2012253760B2 (en) 2011-05-06 2016-02-04 Zafgen, Inc. Tricyclic pyrazole sulfonamide compounds and methods of making and using same
EA025526B1 (ru) 2011-05-06 2017-01-30 Зафджен Инк. Частично насыщенные трициклические соединения и способы их получения и применения
AU2012253759B2 (en) 2011-05-06 2016-01-21 Zafgen Inc. Tricyclic sulfonamide compounds and methods of making and using same
KR101350824B1 (ko) * 2011-12-14 2014-01-13 서울대학교산학협력단 리파아제 특이적 지질-펩타이드 기질 라이브러리 및 이를 이용한 리파아제 저해제를 유효성분으로 함유하는 비만 및 지질대사 이상 관련 질환의 치료 및 예방용 약학적 조성물
US20130158077A1 (en) 2011-12-19 2013-06-20 Ares Trading S.A. Pharmaceutical compositions
US9504679B2 (en) 2011-12-19 2016-11-29 Bjoern Colin Kahrs Pharmaceutical compositions comprising glitazones and Nrf2 activators
US9359369B2 (en) 2012-01-18 2016-06-07 Zafgen, Inc. Tricyclic sulfonamide compounds and methods of making and using same
BR112014017780A8 (pt) 2012-01-18 2017-07-11 Zafgen Inc Compostos de sulfona tricíclicos e métodos para fazer e usar os mesmos
US20150045610A1 (en) * 2012-04-12 2015-02-12 Monell Chemical Senses Center Method of modifying spermatogenesis, spermiogenesis and/or fertility in mammals
US9839622B2 (en) 2012-05-08 2017-12-12 Zafgen, Inc. Methods of treating hypothalamic obesity
WO2013169860A1 (fr) 2012-05-09 2013-11-14 Zafgen, Inc. Composés de type fumagillol, leurs procédés de fabrication et d'utilisation
AU2013337287B2 (en) 2012-11-05 2017-03-23 Zafgen, Inc. Methods of treating liver diseases
CN104870433A (zh) 2012-11-05 2015-08-26 扎夫根股份有限公司 三环化合物及其制备方法和用途
CA2890344A1 (fr) 2012-11-05 2014-05-08 Zafgen, Inc. Composes tricycliques a utiliser dans le traitement et/ou la lutte contre l'obesite
ES2701077T3 (es) * 2012-11-29 2019-02-20 St Pharm Co Ltd Conjugado de oligómero de ácido biliar para un nuevo transporte vesicular y uso del mismo
WO2014108449A1 (fr) 2013-01-08 2014-07-17 Atrogi Ab Procédé de criblage, trousse, méthode de traitement et composé destinés à être utilisé dans une méthode de traitement
CN107501110B (zh) 2013-03-14 2020-05-05 阿尔克梅斯制药爱尔兰有限公司 富马酸酯前体药物及其在治疗多种疾病中的应用
US9597309B2 (en) 2013-03-14 2017-03-21 Zafgen, Inc. Methods of treating renal disease and other disorders
US8669281B1 (en) 2013-03-14 2014-03-11 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US20140275138A1 (en) * 2013-03-15 2014-09-18 Cba Pharma, Inc. Method and products for treating diabetes
EP3024454B1 (fr) 2013-07-25 2020-10-21 MediciNova, Inc. Procédés pour réduire les niveaux de sang de triglycéride, de cholestérol total et de lipoprotéine de basse densité
WO2015120065A1 (fr) * 2014-02-05 2015-08-13 The Trustees Of Columbia University In The City Of New York Inhibition de la gamma-sécrétase réduisant les taux d'apoc3 et les triglycérides du plasma
WO2015127450A1 (fr) 2014-02-24 2015-08-27 Alkermes Pharma Ireland Limited Sulfonamide et promédicaments de fumarates de sulfinamide et leur utilisation dans le traitement de diverses maladies
AR105671A1 (es) 2015-08-11 2017-10-25 Zafgen Inc Compuestos heterocíclicos de fumagillol y sus métodos de elaboración y uso
CN106432255A (zh) 2015-08-11 2017-02-22 扎夫根公司 烟曲霉素醇螺环化合物和制备和使用其的方法
US11266730B2 (en) 2015-09-29 2022-03-08 The General Hospital Corporation Methods of treating and diagnosing disease using biomarkers for BCG therapy
EP3426239A1 (fr) * 2016-03-07 2019-01-16 Atrogi AB Composés pour le traitement de l'hyperglycémie
WO2018204829A1 (fr) * 2017-05-04 2018-11-08 University Of Maryland, Baltimore Procédés pour empêcher des défauts de tube neural dans une grossesse diabétique
GB201714740D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
GB201714734D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
GB201714745D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
GB201714736D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
AU2019315823A1 (en) * 2018-07-31 2021-04-01 Wen Tan New use of carbamate beta phenylethanolamine analogues for enhancing intracellular clearance of ldl cholesterol and for combining therapy with statins to enhance the efficacy and reduce adverse effects
CN113041251B (zh) * 2018-08-31 2022-12-13 中国科学院深圳先进技术研究院 葫芦素e在制备治疗器官移植后血管新生不足药物或生物医用材料中的用途和应用其的产品
CN109913522A (zh) * 2019-04-03 2019-06-21 五邑大学 茶多肽在制备改善、缓解或治疗痛风药物中的应用
EP4149962A1 (fr) * 2020-05-11 2023-03-22 Energesis Pharmaceuticals, Inc. Procédés et compositions pour induire une adipogenèse brune
CA3218880A1 (fr) * 2021-05-20 2022-11-24 Olivier D. Boss Methodes et compositions pour induire une adipogenese brune

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6576256B2 (en) * 2001-08-28 2003-06-10 The Brigham And Women's Hospital, Inc. Treatment of patients at elevated cardiovascular risk with a combination of a cholesterol-lowering agent, an inhibitor of the renin-angiotensin system, and aspirin
WO2003059294A2 (fr) * 2002-01-14 2003-07-24 Pharmacia Corporation Combinaisons d'agonistes du recepteur alpha activateur de la proliferation des peroxisomes et d'inhibiteurs selectifs de la cyclooxygenase-2 et leurs utilisations therapeutiques
WO2003082283A2 (fr) * 2002-03-25 2003-10-09 Jian Luo Compositions pharmaceutiques contenant une combinaison de principes actifs et procedes permettant de les utiliser
WO2004052362A1 (fr) * 2002-12-10 2004-06-24 Novartis Ag Combinaision d'un inhibiteur de dpp-iv et d'un compose ppar-alpha
WO2005025673A1 (fr) * 2003-09-08 2005-03-24 Franco Folli Therapie multisystemique contre le diabete, le syndrome metabolique et l'obesite comprenant un agent hypoglycemique

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA65635C2 (uk) * 1998-09-03 2004-04-15 Н-Гене Кутато Кфт. НЕНАСИЧЕНІ ПОХІДНІ ГІДРОКСИМОВОЇ КИСЛОТИ, ЩО МАЮТЬ ВЛАСТИВОСТІ ІНГІБІТОРІВ NAD<sup>+</sup>-ADP-РИБОЗИЛТРАНСФЕРАЗИ
US6297260B1 (en) * 1998-10-30 2001-10-02 Nitromed, Inc. Nitrosated and nitrosylated nonsteroidal antiinflammatory compounds, compositions and methods of use
ES2234523T3 (es) * 1999-03-01 2005-07-01 Pfizer Products Inc. Acidos oxamicos que contienen ciano y sus derivados como ligandos de receptores tiroideos.
WO2001080896A2 (fr) * 2000-04-21 2001-11-01 Arch Development Corporation Combinaisons medicamenteuses a base de flavopiridol, et methodes correspondantes, aux effets secondaires moindres

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6576256B2 (en) * 2001-08-28 2003-06-10 The Brigham And Women's Hospital, Inc. Treatment of patients at elevated cardiovascular risk with a combination of a cholesterol-lowering agent, an inhibitor of the renin-angiotensin system, and aspirin
WO2003059294A2 (fr) * 2002-01-14 2003-07-24 Pharmacia Corporation Combinaisons d'agonistes du recepteur alpha activateur de la proliferation des peroxisomes et d'inhibiteurs selectifs de la cyclooxygenase-2 et leurs utilisations therapeutiques
WO2003082283A2 (fr) * 2002-03-25 2003-10-09 Jian Luo Compositions pharmaceutiques contenant une combinaison de principes actifs et procedes permettant de les utiliser
WO2004052362A1 (fr) * 2002-12-10 2004-06-24 Novartis Ag Combinaision d'un inhibiteur de dpp-iv et d'un compose ppar-alpha
WO2005025673A1 (fr) * 2003-09-08 2005-03-24 Franco Folli Therapie multisystemique contre le diabete, le syndrome metabolique et l'obesite comprenant un agent hypoglycemique

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
See also references of WO2006004803A1 *
SONE H ET AL: "Ibuprofen-related hypoglycemia in a patient receiving sulfonylurea." ANNALS OF INTERNAL MEDICINE 20 FEB 2001, vol. 134, no. 4, 20 February 2001 (2001-02-20), page 344, XP002479422 ISSN: 0003-4819 *
YUAN M ET AL: "REVERSAL OF OBESITY- AND DIET-INDUCED INSULIN RESISTANCE WITH SALICYLATES OR TARGETED DISRUPTION OF IKKBETA" SCIENCE, WASHINGTON, DC, vol. 293, no. 5535, 31 August 2001 (2001-08-31), pages 1673-1677, XP001018090 ISSN: 0036-8075 *

Also Published As

Publication number Publication date
NO20070510L (no) 2007-03-29
AU2005259864A1 (en) 2006-01-12
IL180251A0 (en) 2007-09-20
TW200608961A (en) 2006-03-16
BRPI0512856A (pt) 2008-04-08
CA2571683A1 (fr) 2006-01-12
JP2008505176A (ja) 2008-02-21
AR049955A1 (es) 2006-09-20
US20060069161A1 (en) 2006-03-30
EP1781303A4 (fr) 2008-07-02
WO2006004803A1 (fr) 2006-01-12
RU2007103178A (ru) 2008-08-10
MX2007000142A (es) 2007-03-26
KR20070027747A (ko) 2007-03-09
US7795310B2 (en) 2010-09-14

Similar Documents

Publication Publication Date Title
US7795310B2 (en) Methods and reagents for the treatment of metabolic disorders
Pierce et al. Heart dysfunction in diabetes
US6204245B1 (en) Treatment of narcolepsy with immunosuppressants
US8377947B2 (en) Treating alzheimer&#39;s disease and osteoporosis and reducing aging
US20050192261A1 (en) Methods and reagents for the treatment of immunoinflammatory disorders
US20070213296A1 (en) Compositions and methods for the treatment of immunoinflammatory disorders
MXPA06013463A (es) Metodos y reactivos para el tratamiento de desordenes inmuno-inflamatorios.
TR201802359T4 (tr) Metabolik sendromun tedavi edilmesine yönelik yeni bileşim.
MX2008012506A (es) Tratamiento de combinacion de trastornos metabolicos.
CA2644933A1 (fr) Combinaisons therapeutiques et methodes pour amelioration cardio-vasculaire et traitement de maladie cardio-vasculaire
CN101010083A (zh) 治疗代谢性病症的方法和试剂
US20040220153A1 (en) Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
US20060281722A1 (en) Compounds and uses thereof
Chan et al. Adipose lipoprotein lipase in insulin-treated diabetic lean and obese Zucker rats
US20150037361A1 (en) Compositions and methods to treat the bihormonal disorder in diabetes
US20230190689A1 (en) Methods and compositions for inducing brown adipogenesis
KR20070022758A (ko) 면역염증성 질환의 치료 방법 및 시약
US20060234910A1 (en) Methods for the treatment of insulin resistance and disease states characterized by insulin resistance
Brawerman Maternal resveratrol supplementation in gestational diabetes prevents cardio-metabolic disease development and improves cardiac structure in the rat offspring
US20100069377A1 (en) Treatment of female sexual dysfunction by compounds that positively modulate ampa-type glutamate receptors
WO2023094692A1 (fr) Traitement de la polyarthrite rhumatoïde
Olson PPAR gamma and PPAR delta in glucose and lipid homeostasis
Flores Orphanin FQ produces sex-specific modulation of trigeminal nociception: Electrophysiological behavioral and molecular studies
Bae et al. Repression of γ-Glutamylcysteine Synthetase and Glutathione S-Transferases by Metformin, an Anti-diabetic Agent, in H4IIE Rat Hepatocytes

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070130

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RAX Requested extension states of the european patent have changed

Extension state: YU

Payment date: 20070130

Extension state: MK

Payment date: 20070130

Extension state: HR

Payment date: 20070130

Extension state: BA

Payment date: 20070130

Extension state: AL

Payment date: 20070130

A4 Supplementary search report drawn up and despatched

Effective date: 20080602

17Q First examination report despatched

Effective date: 20100416

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ZALICUS INC.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ZALICUS INC.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20101228